Carbonic Anhydrase VIII (CA8) is a unique member of the carbonic anhydrase family that lacks traditional carbonic anhydrase enzymatic activity due to the absence of catalytic zinc coordinating residues . Despite its structural similarity to other carbonic anhydrases, CA8 functions as an IP3R1-binding protein (also known as CARP) and is highly expressed in Purkinje cells in the cerebellum . Its significance stems from its regulatory role in calcium signaling - it binds to IP3R1 and reduces the receptor's affinity for its ligand IP3 . CA8 has gained research attention because defects in this protein are associated with cerebellar ataxia mental retardation and dysequilibrium syndrome type 3 (CMARQ3) . The protein consists of 290 amino acids with a calculated molecular weight of approximately 32 kDa, though in some detection methods it can appear at slightly different sizes (36-50 kDa) depending on experimental conditions .
CA8 antibodies have been validated for multiple experimental applications across various research settings. Based on available data, these antibodies are suitable for:
Western Blot (WB): Validated for detection of CA8 in human and mouse cerebellum tissue lysates
Immunohistochemistry (IHC): Successfully used for tissue staining, particularly in brain tissues
Immunofluorescence (IF): Applicable for cellular localization studies
Immunoprecipitation (IP): Useful for protein isolation and interaction studies
Mass Cytometry (CyTOF): Demonstrated positive results in ovarian cancer tissue and various normal tissues
For optimal results, each laboratory should determine appropriate dilutions. For Western blot applications, antibody concentrations ranging from 0.5-5 μg/mL have been successfully employed depending on the specific antibody clone .
Available CA8 antibodies demonstrate varied species reactivity profiles depending on the specific antibody clone and manufacturer. Based on the search results, the following reactivity patterns have been documented:
| Antibody | Confirmed Reactivity | Predicted Reactivity |
|---|---|---|
| ABIN6740749 | Human, Mouse, Rat, Cow, Dog, Zebrafish | Guinea Pig, Horse, Monkey, Rabbit, Bat, Chicken, Pig |
| AF2187 | Human, Mouse | Not specified |
| MAB2187 | Human, Mouse | Not specified |
| 12391-1-AP | Human, Mouse, Rat | Not specified |
| CPTC-CA8-1/2 | Human | Not specified |
The high degree of sequence conservation of CA8 across species (as high as 100% identity in many mammals and 92% in some non-mammalian vertebrates) explains the broad cross-reactivity observed with many antibodies . This makes certain antibodies particularly valuable for comparative studies across multiple model organisms .
CA8 antibodies have been validated for detection in multiple sample types:
Brain tissue lysates: Particularly cerebellum tissue, where CA8 is highly expressed
Cell lysates: From cell lines expressing endogenous or recombinant CA8
Tissue sections: For immunohistochemistry applications in normal and pathological samples
Recombinant protein: For antibody characterization and as positive controls
Notably, CA8 has been successfully detected in various normal tissues beyond the brain, including colon, pancreas, breast, lung, testis, endometrium, and kidney, as well as in cancer tissues (breast, colon, ovarian, lung, and prostate) . For optimal results, proper sample preparation protocols should be followed based on the specific application and antibody being used.
Optimizing Western blot protocols for CA8 detection requires attention to several critical parameters:
Lysate preparation and loading:
For brain tissue lysates (particularly cerebellum), a concentration of 0.2 mg/mL has been successfully used
Include protease inhibitors to prevent degradation of CA8 during sample preparation
Use reducing conditions, as demonstrated in successful detection protocols
Antibody selection and dilution:
For polyclonal antibodies like AF2187, a concentration of 0.5 μg/mL has shown specific detection
For monoclonal antibodies like MAB2187, a concentration of 1 μg/mL has been effective
Detection system optimization:
Use appropriate secondary antibodies: HRP-conjugated Anti-Goat IgG (for AF2187) or Anti-Mouse IgG (for MAB2187)
Immunoblot Buffer Group 1 has been successfully used in published protocols
PVDF membranes have been successfully employed for CA8 detection
Band interpretation:
Expect the main CA8 band at approximately 32-36 kDa in most standard conditions
In Simple Western systems, additional bands at 41 and 50 kDa have been observed
Compare with positive controls (recombinant CA8 protein) to confirm specificity
Following these optimized protocols will help ensure specific detection of CA8 while minimizing background and non-specific signals.
Immunohistochemical detection of CA8 in neural tissues requires careful attention to several factors:
Tissue fixation and preparation:
Optimized fixation protocols are crucial as overfixation may mask epitopes
For CA8 detection in cerebellum, standard formalin fixation followed by paraffin embedding has been successful
Antigen retrieval methods may be necessary and should be optimized
Antibody selection:
Monoclonal antibodies CPTC-CA8-1 and CPTC-CA8-2 have been validated for IHC applications by the Human Protein Atlas
Polyclonal antibody 12391-1-AP has also demonstrated suitability for IHC applications
Signal interpretation:
CA8 shows strong expression in Purkinje cells of the cerebellum
Co-localization studies with IP3R1 may provide additional information about CA8 distribution and function
Compare staining patterns with published references to confirm specificity
Controls:
Include known positive tissues (cerebellum) and negative controls (tissues with minimal CA8 expression)
Consider using genetic models (CA8 knockout or knockdown) as negative controls where available
Include isotype controls to assess non-specific binding
For advanced applications like double immunostaining, additional optimization steps including careful selection of secondary antibodies to avoid cross-reactivity will be necessary.
The choice between monoclonal and polyclonal CA8 antibodies significantly impacts experimental outcomes:
Monoclonal Antibodies (e.g., MAB2187, CPTC-CA8-1/2):
Advantages: Consistent lot-to-lot performance, high specificity for a single epitope, reduced background in some applications
Optimal applications: IHC of specific structures, quantitative analyses, applications requiring high reproducibility
Performance observations: Clone 308320 (MAB2187) effectively detects a specific 36 kDa band in Western blot
CPTC-CA8-1 and CPTC-CA8-2 have shown positive results in IHC evaluations by the Human Protein Atlas
Polyclonal Antibodies (e.g., AF2187, ABIN6740749, 12391-1-AP):
Advantages: Recognition of multiple epitopes, potentially higher sensitivity, better tolerance to protein denaturation
Optimal applications: Western blotting of denatured proteins, detection of proteins present in low abundance
Performance observations: AF2187 detects bands at 36 kDa in standard Western blot and additional bands at 41 and 50 kDa in Simple Western systems
The C-terminal targeted ABIN6740749 offers broad species reactivity
Comparative considerations:
For critical quantitative studies, monoclonals may offer better reproducibility
For maximum sensitivity or detection of CA8 across multiple species, polyclonals may be advantageous
When studying potential post-translational modifications, polyclonals recognizing multiple epitopes may detect more variants
For co-localization studies with IP3R1, antibodies raised against non-binding regions of CA8 may be preferred
Selection should be based on the specific experimental requirements, with consideration for the epitope recognized by each antibody clone.
When investigating CA8 in neurological disorders, particularly cerebellar ataxia-related conditions, implementing robust controls is critical:
Positive controls:
Wild-type cerebellum tissue: Known to express high levels of CA8, particularly in Purkinje cells
Recombinant CA8 protein: Useful for antibody validation and as a standard in quantitative studies
Cell lines with confirmed CA8 expression: Important for validating detection methods
Negative controls:
CA8 knockout or knockdown models: Essential for confirming antibody specificity
Tissues with minimal CA8 expression: To establish detection thresholds
Secondary antibody-only controls: To assess background staining
Disease-specific controls:
Age-matched control samples: Essential when studying neurodegenerative conditions
Samples representing disease progression stages: To track CA8 alterations over time
Samples from related neurological disorders: To identify CA8 changes specific to the disorder of interest
Technical validation controls:
Loading controls for Western blots: To ensure equal protein loading across samples
Housekeeping gene expression: For normalization in transcriptional studies
Multiple antibodies targeting different CA8 epitopes: To confirm consistent detection patterns
Inclusion of these controls will strengthen experimental design and increase confidence in findings related to CA8's role in neurological disorders, particularly those involving cerebellar dysfunction.
Researchers frequently encounter variations in CA8 molecular weight detection across different experimental platforms. Understanding and resolving these discrepancies requires systematic approaches:
Observed variations:
Standard Western blot: CA8 typically detected at approximately 36 kDa
Simple Western system: Additional bands observed at 41 and 50 kDa
Calculated molecular weight: 32 kDa based on amino acid sequence
Potential causes and resolution strategies:
Post-translational modifications:
Phosphorylation, glycosylation, or other modifications may alter migration patterns
Resolution: Use phosphatase or glycosidase treatments to confirm modification status
Compare migration patterns under different reducing conditions
Alternative splicing:
Different CA8 isoforms may exist with varied molecular weights
Resolution: Use isoform-specific primers in RT-PCR to confirm expression of variants
Compare detection patterns across different tissues known to express specific isoforms
Methodological variations:
Different electrophoresis systems and conditions can affect protein migration
Resolution: Run samples on multiple gel types (gradient vs. fixed percentage)
Include recombinant CA8 protein as a migration standard
Antibody-specific detection:
Different antibody clones may recognize distinct forms of CA8
Resolution: Compare detection patterns using multiple antibodies targeting different epitopes
Perform immunoprecipitation followed by mass spectrometry to confirm protein identity
When reporting results, researchers should clearly specify their detection method, antibody used, and observed molecular weight to facilitate meaningful comparison across studies.
Given CA8's functional relationship with IP3R1, optimizing co-immunoprecipitation (co-IP) protocols is essential for advancing understanding of their physiological interactions:
Lysate preparation considerations:
Use gentle lysis buffers to preserve protein-protein interactions (e.g., RIPA buffer with reduced detergent concentration)
Include phosphatase inhibitors, as phosphorylation may influence binding interactions
Prepare lysates from cerebellum tissue or Purkinje cell-enriched cultures where both proteins are abundantly expressed
Antibody selection:
For CA8 pull-down: The 12391-1-AP antibody has been validated for immunoprecipitation applications
Choose antibodies targeting epitopes away from the interaction interface to avoid interference with binding
Confirm antibody specificity in both Western blot and IP applications before co-IP experiments
Technical optimization:
Pre-clear lysates to reduce non-specific binding
Consider crosslinking approaches for transient interactions
Compare native versus crosslinked conditions to assess interaction stability
Include appropriate controls (IgG control, lysates from tissues with low CA8 expression)
Verification approaches:
Reciprocal co-IP (pull down with IP3R1 antibody and detect CA8, and vice versa)
Validate interactions using alternative methods (proximity ligation assay, FRET)
Consider using recombinant tagged proteins for in vitro binding studies to complement co-IP findings
These optimized approaches will help establish the specificity and physiological relevance of CA8-IP3R1 interactions, particularly in the context of cerebellar function and related pathologies.
Non-specific binding and high background are common challenges when working with CA8 antibodies. Implementing the following targeted strategies can significantly improve signal-to-noise ratio:
For Western blot applications:
Increase blocking time or concentration (5% BSA or milk in PBS-T)
Optimize antibody dilution through careful titration experiments
Consider using alternative membranes (PVDF has been successful for CA8 detection)
Include 0.1% BSA in antibody solutions to reduce non-specific binding
Increase washing steps duration and frequency (5-6 washes of 5-10 minutes each)
Pre-adsorb antibodies with non-target tissue lysates when cross-reactivity is observed
For immunohistochemistry/immunofluorescence:
Use tissue-specific blocking solutions containing serum from the same species as the secondary antibody
Implement endogenous peroxidase quenching for HRP-based detection systems
Reduce primary antibody concentration and increase incubation time
For mouse tissue: Consider using mouse-on-mouse blocking kits when using mouse monoclonals
Optimize antigen retrieval conditions (method, pH, duration)
Implement Sudan Black B treatment to reduce autofluorescence in neural tissues
General optimization considerations:
For polyclonal antibodies: Consider affinity purification against the immunogen
Validate antibody specificity using knockout/knockdown controls
Use multiple antibodies targeting different epitopes to confirm staining patterns
Implement careful negative controls (no primary antibody, isotype controls)
These systematic approaches will help distinguish specific CA8 signals from background, particularly in complex neural tissues where CA8 is expressed in specific cell populations.
Distinguishing CA8 from other carbonic anhydrase family members requires careful experimental design due to structural similarities despite functional differences:
Antibody selection strategies:
Choose antibodies raised against unique regions of CA8 that show minimal sequence homology with other family members
C-terminal targeted antibodies like ABIN6740749 may offer improved specificity
Validate antibody specificity against recombinant proteins of multiple CA family members
Perform pre-adsorption controls with recombinant proteins of related CA family members
Expression pattern analysis:
Leverage CA8's distinctive expression pattern (high in cerebellum, particularly Purkinje cells)
Compare with known distribution patterns of other CA family members
Analyze co-expression patterns with cell-type specific markers
Functional discrimination approaches:
Unlike other CA family members, CA8 lacks carbonic anhydrase activity due to absence of catalytic zinc coordinating residues
Design functional assays that distinguish enzymatically active CAs from the inactive CA8
Assess IP3R1 binding capacity, which is characteristic of CA8 but not other CA family members
Molecular techniques for specific detection:
Design primers/probes targeting unique regions for qPCR discrimination
Use CRISPR-Cas9 to specifically tag or modify endogenous CA8
Implement RNA-seq analysis to comprehensively profile expression of all CA family members
These multi-faceted approaches ensure accurate identification of CA8 in experimental systems, preventing misattribution of signals to other carbonic anhydrase family members.
When knockout models are unavailable, researchers can implement alternative validation strategies to confirm CA8 antibody specificity:
RNA interference approaches:
siRNA or shRNA knockdown of CA8 in cell culture models
Compare antibody signal between knockdown and control cells via Western blot or immunocytochemistry
Quantify reduction in signal intensity correlating with knockdown efficiency
Overexpression systems:
Transfect cells with CA8 expression constructs (tagged or untagged)
Compare antibody staining between transfected and non-transfected cells
Use multiple antibodies to confirm consistent detection patterns
Peptide competition assays:
Pre-incubate antibody with excess immunizing peptide or recombinant CA8 protein
Compare signal between blocked and unblocked antibody conditions
Specific signals should be significantly reduced or eliminated
Multi-antibody validation approach:
Compare staining patterns using multiple antibodies targeting different CA8 epitopes
Consistent patterns across multiple antibodies suggest specific detection
For example, compare results from N-terminal vs. C-terminal targeting antibodies
Mass spectrometry verification:
Perform immunoprecipitation with the CA8 antibody
Analyze precipitated proteins by mass spectrometry
Confirm presence of CA8 peptides and assess co-precipitated proteins
Tissue-specific validation:
Compare staining intensity across tissues with known differential CA8 expression
High signal in cerebellum (especially Purkinje cells) with lower signal in other brain regions
Correlate protein detection with mRNA expression data from public databases
These complementary approaches provide robust validation when genetic knockout models are unavailable, ensuring confident interpretation of experimental results.
CA8 antibodies offer valuable tools for investigating cerebellar ataxia, particularly CMARQ3, which is directly linked to CA8 mutations :
Clinical and translational applications:
Immunohistochemical analysis of post-mortem tissues from ataxia patients to assess CA8 expression patterns
Comparison of CA8 distribution between patient and control tissues to identify pathological alterations
Assessment of CA8-IP3R1 co-localization in disease states to determine if interaction is disrupted
Experimental model characterization:
Validation of animal models of CA8-related disorders through protein expression profiling
Characterization of cellular and subcellular CA8 distribution in mutant models
Assessment of developmental expression patterns in normal and pathological states
Therapeutic development applications:
Monitoring CA8 expression changes in response to experimental therapeutics
Evaluating restoration of normal CA8-IP3R1 interactions following treatment
Screening for compounds that stabilize mutant CA8 or restore its function
Methodological approaches:
Multiplexed immunofluorescence with markers of Purkinje cell health
Temporal analysis of CA8 expression during disease progression
Combined analysis of CA8 protein levels and calcium signaling dynamics
These applications position CA8 antibodies as essential tools in understanding the molecular mechanisms underlying cerebellar ataxias and developing targeted therapeutic approaches.
Recent evidence suggests CA8 may have relevance in cancer research, with detection reported in various cancer tissues . When applying CA8 antibodies in cancer studies, researchers should consider:
Tissue-specific expression patterns:
CA8 has been detected in multiple cancer types including breast, colon, ovarian, lung, and prostate cancers
Compare expression between matched normal and tumor tissues from the same patient
Assess correlation with tumor grade, stage, and other clinical parameters
Methodological optimization:
For CyTOF applications, CPTC-CA8-1 has shown positive results at 1:100 dilution of 0.5mg/mL stock
Optimize antigen retrieval for FFPE cancer tissue sections, which may require different conditions than normal tissues
Consider using tissue microarrays for high-throughput screening across multiple cancer types
Functional correlation studies:
Investigate relationship between CA8 expression and calcium signaling in cancer cells
Assess potential correlation with tumor cell proliferation, migration, or invasion
Evaluate association with treatment response or resistance mechanisms
Validation approaches:
Confirm antibody specificity in cancer tissues using multiple antibodies
Correlate protein detection with mRNA expression data from cancer genomics databases
Use cancer cell lines with manipulated CA8 expression as controls
Technical considerations:
Be aware of potential non-specific binding in necrotic tumor regions
Include appropriate positive controls (cerebellum tissue) and negative controls
Consider the impact of tumor heterogeneity on CA8 expression interpretation
These considerations will enhance the reliability and interpretability of CA8 detection in cancer research applications, potentially opening new avenues for understanding calcium signaling dysregulation in tumorigenesis.
CA8's role in cerebellar function makes it a compelling target for developmental neuroscience research. When using CA8 antibodies in this context, researchers should consider:
Developmental expression profiling:
Design experiments to track CA8 expression across different developmental stages
Compare expression patterns between embryonic, postnatal, and adult tissues
Correlate CA8 expression with key developmental milestones in cerebellar maturation
Cell-type specific analysis:
Implement co-labeling with markers of different neural cell types
Focus on Purkinje cell development, where CA8 is predominantly expressed
Assess potential expression in neural progenitors and migrating neurons
Subcellular localization studies:
Use high-resolution imaging to track CA8 subcellular distribution during development
Monitor co-localization with IP3R1 throughout developmental stages
Assess potential changes in localization during neuronal maturation
Functional correlation approaches:
Correlate CA8 expression with calcium signaling development
Investigate relationship between CA8 expression and dendritic arborization in Purkinje cells
Assess impact of CA8 manipulation on developmental milestones
Technical optimization:
Adapt fixation and permeabilization protocols for embryonic and early postnatal tissues
Implement tissue clearing techniques for 3D reconstruction of expression patterns
Consider the higher cellular density of developing tissues when optimizing antibody dilutions
These approaches will advance understanding of CA8's role in neurodevelopment, potentially revealing insights into how its dysfunction contributes to developmental neurological disorders.
Various detection methods offer distinct advantages for CA8 analysis, with important considerations for selecting the optimal approach:
Key considerations for method selection:
For basic expression studies: Standard Western blot offers reliable detection with established protocols
For detailed localization: IHC or IF provides cellular and subcellular resolution
For interaction studies: IP or proximity ligation assays are optimal for CA8-IP3R1 binding
For multiplex analysis: CyTOF allows simultaneous detection of CA8 with multiple markers
Method selection should be guided by the specific research question, available sample types, and required sensitivity and specificity thresholds.
While antibodies are valuable tools for CA8 detection, combining them with complementary approaches provides deeper functional insights:
Genetic manipulation approaches:
CRISPR-Cas9 gene editing to introduce specific CA8 mutations
Conditional knockout models to study cell-type specific functions
Targeted knock-in of tagged CA8 for live imaging studies
Expression of mutant forms associated with CMARQ3 for pathophysiological studies
Advanced imaging techniques:
FRET-based assays to monitor CA8-IP3R1 interaction dynamics in real-time
Calcium imaging to assess the impact of CA8 manipulation on IP3-mediated calcium signaling
Super-resolution microscopy for nanoscale localization of CA8 relative to IP3R1
Live-cell imaging with tagged CA8 to track trafficking and interaction dynamics
Biochemical and structural approaches:
Surface plasmon resonance to quantify CA8-IP3R1 binding kinetics
Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces
X-ray crystallography or cryo-EM of CA8-IP3R1 complexes
In vitro reconstitution assays to assess direct functional impacts on calcium signaling
Systems biology approaches:
Proteomics analysis of CA8 interactome beyond IP3R1
Transcriptomics following CA8 manipulation to identify downstream pathways
Computational modeling of CA8's impact on calcium signaling networks
Multi-omics integration to place CA8 in broader cerebellar function context
These complementary approaches, when combined with traditional antibody applications, provide a comprehensive understanding of CA8's functional roles in normal physiology and disease states.
Comprehensive validation of novel CA8 antibodies, particularly for specialized applications, requires systematic multi-step approaches:
Initial characterization:
ELISA against immunizing peptide/protein to confirm binding
Western blot using recombinant CA8 protein to confirm specificity and determine sensitivity
Comparison with existing validated antibodies using the same detection methods
Cross-reactivity testing against related carbonic anhydrase family members
Application-specific validation:
For Western blot: Test across multiple sample types including cerebellum tissue and cell lines
For IHC/IF: Validate in tissues with known CA8 expression patterns, focusing on Purkinje cells
For IP: Confirm ability to precipitate CA8 and co-precipitate known binding partners like IP3R1
For mass cytometry: Validate metal conjugation efficiency and epitope accessibility
Stringent specificity controls:
Test in CA8 knockout/knockdown models if available
Perform peptide competition assays
Compare staining patterns with in situ hybridization data
Validate across multiple species if cross-reactivity is claimed
Performance documentation:
Determine optimal working dilutions for each application
Establish detection limits and linear range for quantitative applications
Document lot-to-lot variation for polyclonal antibodies
Assess storage stability and freeze-thaw tolerance
Advanced validation for specialized applications:
For proximity ligation assays: Validate with known interaction partners
For multiplexed imaging: Test for antibody cross-talk with other detection channels
For live cell applications: Confirm epitope accessibility in non-fixed conditions
For phospho-specific detection: Validate with phosphatase treatments and phosphomimetic mutants
Thorough validation following these guidelines ensures reliable performance in specialized research applications and supports reproducible CA8 research outcomes.
CA8 antibodies hold significant potential for advancing personalized approaches to cerebellar disorders, particularly CA8-associated CMARQ3 and related conditions:
Diagnostic applications:
Development of antibodies specific to common CA8 mutations
Implementation in diagnostic panels for ataxia and related movement disorders
Correlation of CA8 expression patterns with clinical phenotypes and disease progression
Pharmacodynamic biomarker development:
Monitoring CA8 protein levels in accessible samples (CSF, exosomes) during clinical trials
Assessing restoration of normal CA8-IP3R1 interaction as a treatment response marker
Development of phospho-specific antibodies to track CA8 activation state during treatment
Therapeutic stratification:
Identification of patient subgroups based on CA8 expression patterns
Correlation of CA8 variants with response to calcium signaling modulators
Prediction of treatment outcomes based on CA8 functional status
Novel therapeutic development:
Therapeutic antibody development targeting function-modulating CA8 epitopes
Screening compounds that stabilize mutant CA8 protein
Identification of small molecules that modulate CA8-IP3R1 interaction
Implementation considerations:
Development of standardized clinical assays with consistent sensitivity/specificity
Creation of reference standards for quantitative CA8 detection
Validation across diverse patient populations