CAPN5 (Calpain-5) is a calcium-dependent cysteine protease involved in proteolysis and signal transduction pathways. In humans, the canonical protein consists of 640 amino acid residues with a molecular mass of approximately 73.2 kDa . As a member of the Peptidase C2 protein family, CAPN5 differs from classical calpains (CAPN1 and CAPN2) by containing a C2-like domain instead of a penta-EF hand domain at its C-terminus .
Functionally, CAPN5:
Participates in calcium-regulated non-lysosomal proteolysis
Interacts with cytoskeletal proteins
Promotes degradation or remodeling that facilitates normal cellular function
Associates with promyelocytic leukemia protein bodies (PML) in the nucleus, which are implicated in cellular stress response, apoptosis, cellular senescence, and protein degradation
CAPN5 demonstrates a varied expression pattern across tissues and species:
In humans:
Expressed in numerous tissues throughout the body
Represents the second most abundantly expressed calpain in the central nervous system
In retinal tissue, specifically localizes to:
Across species:
CAPN5 orthologs have been identified in multiple species including:
The zebrafish has two CAPN5 orthologs (capn5a and capn5b) with capn5a showing 68% identity and 81% similarity to human CAPN5, while capn5b shows 71% identity and 83% similarity .
The primary disease associated with CAPN5 mutations is Autosomal Dominant Neovascular Inflammatory Vitreoretinopathy (ADNIV), a devastating inherited autoimmune disease of the eye (OMIM #193235) . ADNIV presents as a bilateral panuveitis and displays features commonly seen in other eye diseases, including:
Disease mechanism:
ADNIV results from gain-of-function mutations in CAPN5 that lower the calcium threshold required for activation, resulting in hyperactivity of the protease . Key mutations include:
| Mutation | Location | Effect | Phenotype Severity |
|---|---|---|---|
| p.Arg243Leu (c.728G>T) | Proteolytic core (PC2) | Mislocalization from membrane to cytosol | Moderate |
| p.Leu244Pro (c.731T>C) | Proteolytic core (PC2) | Mislocalization from membrane to cytosol | Moderate |
| p.Lys250Asn (c.750G>T) | Proteolytic core (PC2) | Mislocalization from membrane to cytosol | Moderate |
| p.Gly267Ser (с.799G>A) | Proteolytic core (PC2) | Unknown | Moderate |
| p.Arg289Trp (c.865C>T) | Proteolytic core (PC2) | Disrupts calcium-dependent regulation | Severe (includes hearing loss and developmental delay) |
| p.Gly376Ser (c.1126G>A) | Domain III (CBSW) | Unknown | Mild |
These mutations are located within the calcium-sensitive domain 2 near the active site and are thought to cause the mislocalization of CAPN5 from the cell membrane to the cytosol .
Loss-of-function (LOF) research has provided critical insights for potential therapeutic approaches targeting CAPN5:
Key findings from genetic inactivation studies:
Photoreceptor-specific knockout (KO) mice for Capn5 showed no significant retinal abnormalities when examined by histology and electroretinography
Genetic databases from 60,706 unrelated subjects without severe disease phenotypes revealed 22 LOF CAPN5 variants located throughout the gene and in all major protein domains
LOF variants were found near known disease-causing variants within the proteolytic core and in regions of high homology between human CAPN5 and 150 homologs
Therapeutic implications:
These findings support that:
Localized inhibition of CAPN5 is a viable strategy for treating hyperactivating disease alleles
Small molecule pharmacological inhibition of dominant CAPN5 mutations represents a feasible treatment approach that doesn't require development of individual, patient-specific gene therapy vectors
The lack of adverse effects from CAPN5 LOF suggests that CAPN5 inhibition, even below normal biological activity levels, could be an efficacious strategy for future clinical trials
When selecting CAPN5 antibodies for research, consider these critical factors:
Application compatibility:
Epitope recognition:
For full-length protein detection: antibodies targeting conserved domains
For specific isoform detection: antibodies targeting unique regions
Common immunogens include:
Species reactivity:
Verify cross-reactivity with your experimental species. Many commercially available antibodies react with:
Clone selection:
For monoclonal antibodies, specific clones have been validated for particular applications:
Before employing CAPN5 antibodies in complex experiments, comprehensive validation is critical:
1. Specificity validation:
Western blot analysis comparing wild-type samples with:
Peptide competition assays to confirm epitope specificity
Cross-reactivity assessment if working with multiple species
2. Sensitivity assessment:
Determine lower detection limits using serial dilutions of recombinant CAPN5 protein
Test antibody performance on endogenous CAPN5 across different tissue types, especially those with varying expression levels (brain tissue shows high expression)
3. Reproducibility testing:
Perform replicate experiments under identical conditions
Test lot-to-lot consistency if using multiple antibody batches
4. Application-specific validation:
For IHC: Include proper controls (e.g., CAPN5-positive tissues like colon and prostate carcinoma)
For IF: Validate subcellular localization patterns (e.g., photoreceptor inner segments and synaptic terminals in retinal sections)
For WB: Confirm expected molecular weight (73kDa canonical form, though observed range can be 65-73kDa)
5. Functional validation:
Compare antibody detection with gene expression data
Verify antibody recognition of mutant CAPN5 forms if studying disease variants
Optimizing CAPN5 immunohistochemistry for retinal tissue requires special considerations:
Tissue preparation protocol:
Fix retinal tissues in 4% paraformaldehyde for 24 hours
Paraffin-embed tissues following standard protocols
Section tissues at 5-7μm thickness
For antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes
Allow sections to cool at room temperature for 20 minutes
Antibody protocol optimization:
Block non-specific binding with 10% normal serum (matched to secondary antibody host) in PBS with 0.3% Triton X-100
Incubate with primary anti-CAPN5 antibody:
Wash 3× in PBS
Apply appropriate HRP-conjugated secondary antibody for 1 hour at room temperature
Develop using DAB and counterstain with hematoxylin
Special considerations for retinal tissues:
Retinal autofluorescence can interfere with immunofluorescence detection; Sudan Black B (0.1% in 70% ethanol) treatment for 5 minutes after secondary antibody incubation can reduce autofluorescence
For dual labeling with retinal cell markers, use:
Rhodopsin for rod photoreceptors
Cone arrestin for cone photoreceptors
Glutamine synthetase for Müller glia
RBPMS for retinal ganglion cells
Expected localization patterns:
In normal retina, CAPN5 should localize to:
Photoreceptor inner segments
Outer plexiform layer (photoreceptor synaptic terminals)
Some ganglion cells
For optimal Western blot detection of CAPN5:
Sample preparation:
Extract proteins using RIPA buffer supplemented with:
Protease inhibitor cocktail
1mM PMSF
1mM sodium orthovanadate
1mM sodium fluoride
Determine protein concentration using Bradford or BCA assay
Load 5-30μg of total protein per lane (tissue-dependent)
Protocol optimization:
Gel electrophoresis:
Transfer conditions:
Use PVDF membrane (0.45μm pore size)
Transfer at 100V for 1 hour or 30V overnight at 4°C
Blocking and antibody incubation:
Detection:
Use HRP-conjugated secondary antibody (1:5000-1:10000)
Develop using enhanced chemiluminescence (ECL)
Expected results:
Canonical CAPN5 band at approximately 73 kDa
May detect additional bands due to post-translational modifications or proteolytic processing
Troubleshooting guidance:
High background: Increase blocking time or use different blocking agent (BSA instead of milk)
No signal: Verify lysate quality, increase protein loading, or reduce antibody dilution
Multiple bands: Validate specificity with knockout/knockdown controls or peptide competition
CAPN5 antibodies provide valuable tools for retinal disease research:
Disease progression monitoring:
Comparative immunohistochemistry across disease stages:
Proteomics applications:
Therapeutic development:
Target validation:
Use antibodies to verify knockdown efficacy in siRNA/shRNA approaches
Confirm CRISPR-mediated gene editing outcomes
Monitor CAPN5 expression in gene therapy models
Pharmacodynamic marker:
Apply CAPN5 antibodies to assess impact of small molecule inhibitors on:
Protein expression levels
Subcellular localization
Protein-protein interactions
Regenerative medicine applications:
Clinical translation potential:
CAPN5 antibodies can help evaluate the efficacy of localized CAPN5 inhibition strategies
Enable monitoring of treatment response in preclinical models prior to human trials
Investigating CAPN5 regulation requires sophisticated experimental approaches:
Calcium-dependent activation studies:
Use calcium imaging techniques (e.g., Fura-2 AM) in combination with immunofluorescence to correlate calcium fluctuations with CAPN5 activation
Employ calpain activity assays alongside CAPN5 immunoprecipitation to distinguish CAPN5 activity from other calpains
Examine structural changes using:
Fluorescence resonance energy transfer (FRET) sensors
Conformational antibodies that recognize active vs. inactive CAPN5 forms
Post-translational modification analysis:
Combine CAPN5 immunoprecipitation with:
Phospho-specific antibodies
Ubiquitin detection antibodies
Mass spectrometry to identify specific modification sites
Study how disease-causing mutations affect:
The proteolytic core structure
Calcium binding capacity
Activation thresholds
Subcellular localization dynamics:
Use confocal microscopy with CAPN5 antibodies alongside organelle markers to track:
Employ live-cell imaging with GFP-tagged CAPN5 validated against antibody staining patterns
Genetic approaches for regulatory mechanism studies:
CRISPR-Cas9 editing to:
Introduce disease-specific mutations (p.Arg243Leu, p.Leu244Pro, etc.)
Create reporter constructs at endogenous loci
Conditional knockout/knockin systems to study:
Developmental timing of CAPN5 function
Tissue-specific regulation
Age-dependent changes in expression and function
Translational research applications:
Patient-derived cells (e.g., iPSCs differentiated to retinal organoids) immunostained for CAPN5
Zebrafish models with fluorescent reporters validated against antibody staining to visualize real-time regulation in vivo
Advanced proteomic methodologies offer new opportunities for CAPN5 research:
Proximity-dependent labeling techniques:
BioID or APEX2 fusion with CAPN5 to identify proximal proteins in:
Normal vs. disease states
Different subcellular compartments
Response to calcium fluctuation
Validate protein interactions using CAPN5 antibodies for co-immunoprecipitation and immunofluorescence co-localization
Temporal proteomic approaches:
Pulse-chase experiments with CAPN5 antibody pulldown to track:
CAPN5 substrate degradation kinetics
Protein complex formation and disassociation
Time-course analysis of proteomic changes during:
Acute inflammatory responses
Progressive stages of ADNIV pathology
Treatment with anti-inflammatory agents
Single-cell proteomics integration:
Combine CAPN5 antibody-based immunohistochemistry with:
Imaging mass cytometry for multiplexed protein detection
Digital spatial profiling to correlate CAPN5 with inflammatory markers
Integrate findings with single-cell transcriptomics data to create comprehensive models of CAPN5 regulation
Vitreous proteome analysis:
The vitreous proteome of CAPN5-NIV patients has revealed pathway alterations including:
Inflammatory mediators of the acute phase response
Complement cascade activation
These pathways share characteristics with:
Non-infectious posterior uveitis
Rhegmatogenous retinal detachment (RRD)
Age-related macular degeneration (AMD)
Proliferative diabetic retinopathy (PDR)
Zebrafish models offer unique advantages for CAPN5 research and retinal regeneration studies:
Developmental expression profiling:
Whole-mount immunohistochemistry with CAPN5 antibodies to:
Key developmental findings:
Photoreceptor-specific functions:
CAPN5 antibody immunohistochemistry in zebrafish has revealed:
Regeneration research applications:
CAPN5 antibody staining in injury models has shown:
Experimental approaches using CAPN5 antibodies:
Track expression changes during different phases of regeneration
Identify cell types expressing CAPN5 during regenerative response
Compare with mammalian models to identify key differences in regenerative capacity
Translational potential:
Identify pathways where CAPN5 influences regenerative capacity
Develop drug screening platforms in zebrafish to test CAPN5 modulators
Create transgenic zebrafish with fluorescent reporters validated against antibody staining to monitor real-time CAPN5 dynamics during regeneration