Released from neutrophil and eosinophil granules upon activation, suggesting a role in pathogen defense .
Binds α1B-glycoprotein (A1BG) in plasma, forming a 1:1 noncovalent complex to neutralize potential cytotoxic effects .
Expressed in seminal fluid and epididymis, where it associates with β-microseminoprotein (PSP94) to regulate sperm function .
Not detected in the male genital tract of mice, highlighting species-specific roles .
The C-terminal domain modulates ion flux, potentially influencing cellular signaling in inflammatory or cancerous microenvironments .
Upregulation: Strongly overexpressed in prostate cancer compared to normal tissue .
Prognostic Marker: Associated with advanced tumor stage (pT3/pT4), high Gleason score (≥8), and early biochemical recurrence .
Mechanism: Enhances cell motility and invasion by altering cell-cell adhesion proteins (e.g., LASP1, TJP1) .
Biomarker Potential: Plasma CRISP3 levels correlate with sepsis risk in trauma patients:
Outperforms traditional markers like CRP and PCT in sepsis prediction .
Elevated in chronic pancreatitis and severe dengue, linked to tissue remodeling and immune dysregulation .
ELISA Kits: Detect CRISP3 in plasma, serum, and exocrine secretions (sensitivity: ~1.053 ng/mL) .
Immunohistochemistry: Used to localize CRISP3 in cancer tissues .
Human CRISP3 belongs to the CRISP (Cysteine-Rich Secretory Protein) family characterized by 16 invariant cysteine residues forming eight disulfide bonds that define this protein class. It is primarily expressed in exocrine secretions and granulocytes, with significant production in salivary, pancreas, prostate, and lacrimal glands. In the male reproductive system, CRISP3 is expressed in spermatozoa and mature spermatids . Unlike in mice where CRISP3 is not detected in the male genital tract, human CRISP3 shows strong expression in these tissues . The protein appears to play important roles in both reproductive functions and innate immunity responses .
Human CRISP3 exhibits a characteristic three-domain structure:
An N-terminal SCP (Sperm Coating Protein) domain
A hinge region in the middle
The protein contains eight disulfide bonds formed by 16 invariant cysteine residues that are critical for proper folding and structural integrity . Human CRISP3 exists in both glycosylated (30 kDa) and non-glycosylated (28 kDa) forms . The mature protein spans from amino acid residues Asn21 to Tyr245, with researchers often introducing a Ser134Ala mutation in recombinant constructs to improve expression efficiency . When correctly folded, human CRISP3 functions as a monomer in solution, as verified through quaternary structure analysis .
CRISP3 dysregulation has been linked to several pathological conditions. Most notably, CRISP3 is significantly up-regulated in epithelial prostate cancer, making it a potential biomarker for this malignancy . Additionally, increased CRISP3 expression has been observed in chronic pancreatitis . These associations suggest that CRISP3 may serve as both a diagnostic marker and a potential therapeutic target in these conditions. Research into the mechanistic roles of CRISP3 in these pathologies remains an active area of investigation, with particular focus on how its functions in innate immunity and cellular signaling might contribute to disease processes.
While all CRISP proteins share the characteristic 16 conserved cysteine residues, human CRISP3 differs from other family members in several important aspects:
Tissue distribution - CRISP3 has a broader expression pattern compared to other CRISPs, being found in multiple exocrine glands and immune cells .
Glycosylation patterns - Human CRISP3 shows distinct N-glycosylation characteristics that differ from other family members, which impacts its functional properties .
Immunological roles - While many CRISPs in non-mammalian vertebrates function as toxins through ion channel inhibition, human CRISP3 appears more involved in innate immunity processes .
Evolutionary conservation - Interspecies comparison studies reveal interesting differences in CRISP3 structure and glycosylation between humans and mice, suggesting species-specific functional adaptations .
Producing correctly folded human CRISP3 with its eight disulfide bonds presents significant challenges. Bacterial expression systems have shown limited success, yielding low quantities of correctly folded protein . For functional studies requiring authentic human CRISP3, the recommended approach is mammalian expression using HEK 293 cells, which enables:
Proper folding of all eight disulfide bonds
Native glycosylation patterns
Secretion of the protein into culture medium
The purification protocol involves:
Collection of conditioned medium from transfected HEK 293 cells
Initial capture using ion exchange chromatography
Further purification via size exclusion chromatography
Validation of functional authenticity through substrate-affinity assays
This approach yields glycosylated CRISP3 that demonstrates authentic biological activity, making it suitable for downstream functional studies. For applications where glycosylation is not required, researchers may consider using the recombinant protein with a C-terminal 6-His tag, reconstituted from lyophilized form in sterile PBS at 100 μg/mL .
Glycosylation analysis of human CRISP3 requires a multi-method approach:
Mass Spectrometry Analysis:
LC-MS/MS following tryptic digestion to identify glycosylation sites
MALDI-TOF analysis to determine glycan compositions
Enzymatic Deglycosylation Studies:
Treatment with PNGase F to remove N-linked glycans
Treatment with O-glycosidase for O-linked glycan analysis
Comparative SDS-PAGE analysis before and after enzyme treatment
Lectin-Based Glycan Profiling:
Using specific lectins to identify particular glycan structures
Lectin blotting to visualize glycosylation patterns
Research has revealed interesting interspecies differences in CRISP3 glycosylation patterns between humans and mice, underscoring the importance of species-specific analysis . When examining these patterns, researchers should pay particular attention to the 30 kDa (glycosylated) versus 28 kDa (non-glycosylated) forms of the protein .
Given that some CRISP family members in non-mammalian species function through ion channel inhibition, investigating potential interactions between human CRISP3 and ion channels requires specialized techniques:
Patch-Clamp Electrophysiology:
Whole-cell configuration to measure channel currents
Application of purified human CRISP3 to detect modulation of channel activity
Dose-response analyses to determine potency of interaction
Surface Plasmon Resonance (SPR):
Immobilization of purified ion channels on sensor chips
Measurement of binding kinetics with native human CRISP3
Determination of binding affinities and association/dissociation rates
Fluorescence-Based Assays:
Calcium influx measurements in cells expressing specific ion channels
Membrane potential-sensitive fluorescent dyes to detect channel modulation
High-throughput screening of CRISP3 effects on multiple channel types
When designing these experiments, researchers should consider using both the glycosylated and non-glycosylated forms of human CRISP3, as the glycosylation state may influence protein-channel interactions .
The potential role of human CRISP3 in innate immunity, similar to that proposed for equine CRISP3 in inhibiting sperm-neutrophil interactions , can be investigated through:
Neutrophil Interaction Assays:
Co-culture experiments with neutrophils and target cells
Quantification of neutrophil adhesion and activation markers
Assessment of human CRISP3 effects on neutrophil extracellular trap (NET) formation
Cytokine Modulation Studies:
Measurement of pro- and anti-inflammatory cytokine production
RNA-seq or qPCR analysis of immune response genes
Protein array analysis of secreted immune factors
In Vivo Models:
Creation of CRISP3 knockout or transgenic mouse models
Challenge studies with inflammatory stimuli
Assessment of tissue-specific immune responses
These methodologies allow researchers to determine whether human CRISP3 functions similar to its equine ortholog in immunomodulation or if it has species-specific functions in the immune response .
To explore CRISP3's role in male reproductive function, researchers can employ:
Sperm Functional Assays:
Capacitation assessment using chlortetracycline staining
Acrosome reaction quantification with fluorescent lectin binding
Computer-assisted sperm analysis (CASA) for motility parameters
Zona pellucida binding assays to assess fertilization competence
Localization Studies:
Immunofluorescence microscopy to determine subcellular distribution
Immunogold electron microscopy for high-resolution localization
Protein fractionation studies of sperm compartments
Functional Blocking Experiments:
Application of anti-CRISP3 antibodies to block native protein function
Recombinant CRISP3 domain-specific peptides as competitive inhibitors
Assessment of fertilization outcomes following manipulation
These approaches can help determine whether human CRISP3 participates in processes such as decapacitation, acrosome reaction, sperm-oocyte fusion, or flagellar motility, as has been proposed for other CRISP family members .
Accurate quantification of human CRISP3 in various biological samples requires a combination of techniques:
Protein Level Quantification:
Enzyme-linked immunosorbent assay (ELISA) using specific anti-CRISP3 antibodies
Western blotting with densitometric analysis
Mass spectrometry-based quantification (MS/MS with isotope-labeled standards)
mRNA Level Quantification:
Quantitative real-time PCR (qRT-PCR) with validated reference genes
RNA-seq analysis with appropriate normalization
Northern blotting for specific tissue expression patterns
Tissue Expression Analysis:
Immunohistochemistry with specific antibodies
RNA in situ hybridization
Laser capture microdissection combined with qPCR or proteomics
When analyzing CRISP3 expression, researchers should consider both the glycosylated (30 kDa) and non-glycosylated (28 kDa) forms, as their ratio may vary across different tissues and pathological conditions .
Structure-function studies of human CRISP3 require systematic approaches:
Domain Deletion/Mutation Analysis:
Generation of constructs lacking specific domains (SCP domain, hinge region, or cysteine-rich domain)
Site-directed mutagenesis of conserved cysteine residues
Expression in mammalian systems to ensure proper folding
Chimeric Protein Approaches:
Creation of chimeric proteins with domains from other CRISP family members
Functional testing to map domain-specific activities
Structural analysis of chimeric proteins to ensure proper folding
Glycosylation Site Manipulation:
Mutagenesis of N-glycosylation sites (Asn to Gln substitutions)
Comparison of glycosylated versus non-glycosylated variants
Analysis of glycan contribution to stability and function
These approaches should be complemented with structural analysis techniques such as X-ray crystallography or cryo-electron microscopy to correlate functional changes with structural alterations .
When faced with contradictory findings about human CRISP3 function, researchers should consider:
Species-Specific Differences:
Protein Preparation Methods:
Experimental Context Variations:
In vitro versus in vivo experimental settings
Cell type-specific effects that may vary between studies
Concentration-dependent effects that may differ across experimental designs
Methodological Differences:
Antibody specificity issues in immunological detection methods
Variations in recombinant protein tags and their potential interference
Different assay sensitivities and dynamic ranges
To resolve contradictions, researchers should design experiments that directly compare different protein preparations under identical conditions and consider collaborative cross-laboratory validation studies.
Given CRISP3's up-regulation in epithelial prostate cancer , several approaches show promise for biomarker development:
Multi-marker Panel Development:
Integration of CRISP3 with established markers like PSA
Machine learning algorithms to optimize diagnostic accuracy
Longitudinal studies correlating CRISP3 levels with disease progression
Liquid Biopsy Applications:
Detection of CRISP3 in circulating tumor cells
Analysis of CRISP3 in extracellular vesicles from plasma
Development of highly sensitive assays for blood-based detection
Tissue-Based Diagnostic Applications:
Immunohistochemical scoring systems for CRISP3 in prostate biopsies
Correlation with Gleason scores and other histopathological parameters
Development of automated image analysis algorithms for quantification
These approaches should be validated in large, diverse patient cohorts with appropriate controls and follow-up data to establish clinical utility.
Investigating CRISP3 as a therapeutic target requires multi-faceted approaches:
Target Validation Strategies:
CRISPR/Cas9-mediated knockout in relevant cell lines
Inducible expression systems to control CRISP3 levels
In vivo models with tissue-specific CRISP3 modulation
Therapeutic Modulation Approaches:
Development of neutralizing antibodies against specific CRISP3 domains
Small molecule inhibitors targeting protein-protein interactions
RNA interference strategies for expression knockdown
Delivery System Development:
Nanoparticle-based delivery of CRISP3 modulators
Tissue-targeted delivery strategies
Controlled release formulations for sustained effect
When designing these studies, researchers should consider potential compensatory mechanisms from other CRISP family members and carefully assess off-target effects of any therapeutic intervention.
Single-cell approaches offer unprecedented opportunities to elucidate CRISP3 biology:
Single-Cell RNA Sequencing:
Identification of specific cell types expressing CRISP3 within heterogeneous tissues
Analysis of co-expression patterns with potential interaction partners
Trajectory analysis to understand CRISP3 expression during cellular differentiation
Single-Cell Proteomics:
Protein-level confirmation of CRISP3 expression patterns
Post-translational modification analysis at single-cell resolution
Correlation of CRISP3 protein levels with cellular phenotypes
Spatial Transcriptomics/Proteomics:
Mapping CRISP3 expression within tissue microenvironments
Analysis of spatial relationships between CRISP3-expressing cells
Correlation with disease-specific tissue architecture changes
These technologies can help resolve current contradictions in the literature and provide context-specific insights into CRISP3 function across different physiological and pathological states.
Cross-species studies require careful methodological considerations:
Ortholog Selection Criteria:
Sequence homology analysis between species
Phylogenetic mapping to identify true orthologs
Consideration of species-specific gene duplications
Expression System Standardization:
Use of identical expression systems for proteins from different species
Validation of comparable folding and post-translational modifications
Functional testing under identical experimental conditions
Functional Assay Adaptation:
Species-appropriate cellular models
Consideration of species-specific interaction partners
Adaptation of assays to account for biochemical differences
These studies are particularly important given the noted differences between human and mouse CRISP3, including the absence of CRISP3 in the mouse male genital tract compared to its presence in humans .
CRISP-3 is expressed in various tissues, including the male and female reproductive tracts, where it plays roles in sperm function, fertilization, and endometrial receptivity for embryo implantation . It is also found in mammalian exocrine secretions and granulocytes, suggesting a role in innate immunity .