CA8 Human is a recombinant protein produced in E. coli, consisting of 314 amino acids (residues 1–290) with an N-terminal His-Tag. Its molecular mass is 35.5 kDa, and it is purified to >90% purity via chromatographic techniques .
CA8 regulates intracellular calcium signaling by inhibiting ITPR1, a receptor pivotal for endoplasmic reticulum calcium release . This interaction is essential for:
Neuronal Excitability: Modulates Kv7 potassium channels to reduce nociceptor activity, offering potential analgesic effects .
Cerebellar Function: Critical for Purkinje cell development; mutations cause cerebellar ataxia and dysequilibrium syndromes .
CAMRQ3 Syndrome: Autosomal recessive mutations in CA8 lead to cerebellar ataxia, mental retardation, and quadrupedal gait .
Animal Models: CA8-null mice exhibit lifelong gait disorders, mirroring human phenotypes .
Overexpression: Linked to colorectal, lung, and other cancers, though its oncogenic mechanism remains unclear .
Recent studies highlight CA8’s potential in non-opioid analgesia:
Gene Therapy: HSV-based delivery of V5-CA8 in dorsal root ganglia (DRG) reduces neuronal excitability via Kv7 activation, alleviating chronic pain without motor side effects .
Mechanism: Prolongs afterhyperpolarization (AHP) in nociceptors, validated by Kv7 antagonist XE-991 .
CA8 (Carbonic Anhydrase 8) is an atypical member of the carbonic anhydrase family that lacks carbonic anhydrase enzymatic activity due to critical substitutions in its active site . Unlike other family members that catalyze the reversible hydration of carbon dioxide, CA8 functions as an allosteric inhibitor of inositol trisphosphate receptor-1 (ITPR1), regulating neuronal intracellular calcium release . This crucial distinction explains why CA8 is categorized as a "non-classical" carbonic anhydrase.
Methodologically, researchers investigating CA8 should employ techniques focusing on protein-protein interactions and calcium signaling rather than enzymatic activity assays typically used for other carbonic anhydrases. Appropriate approaches include co-immunoprecipitation for detecting CA8-ITPR1 interactions and calcium imaging to assess functional outcomes of CA8 modulation.
Quantitative measurement of human CA8 requires careful selection of methodology based on sample type:
For serum, plasma, and cell culture supernatants:
For tissue and cell lysates:
Western blotting with specific anti-CA8 antibodies allows semi-quantitative assessment
Immunohistochemistry enables visualization of spatial expression patterns
qRT-PCR for mRNA expression analysis as a complementary approach
When designing experiments, researchers should include appropriate controls and validate antibody specificity against recombinant CA8 standards or CA8-knockout samples to ensure accurate results.
Several genetic variants of CA8 have been identified with functional consequences:
Variant | Functional Impact | Associated Phenotypes | Detection Method |
---|---|---|---|
S100P mutation | Loss of ITPR1 inhibition | Increased calcium signaling, altered pain processing | Genotyping, sequencing |
19bp deletion (exon 8) | Loss of function (in mouse Car8) | Ataxia, tremor, calcium dysregulation | PCR, sequencing |
Wild-type | Normal inhibition of ITPR1 | Normal calcium homeostasis | Reference standard |
The S100P mutation represents a particularly important variant that abolishes CA8's ability to inhibit ITPR1-mediated calcium signaling . Researchers should consider genotyping study populations for known CA8 variants, particularly when investigating neurological phenotypes or calcium signaling abnormalities.
When investigating CA8's role in calcium signaling, a comprehensive experimental design should include:
Calcium imaging protocols:
Use ratiometric calcium indicators (Fura-2) or genetically encoded calcium indicators (GCaMP)
Establish baseline calcium levels before stimulation
Apply specific ITPR1 agonists (IP3, ATP) to trigger calcium release
Compare responses between CA8 wild-type, knockout, and overexpression conditions
Molecular interaction studies:
Co-immunoprecipitation to confirm CA8-ITPR1 physical interaction
Proximity ligation assays for visualizing interactions in situ
FRET-based approaches to detect real-time molecular interactions
Phosphorylation analysis:
Western blotting with phospho-specific antibodies to assess ITPR1 and TrkA phosphorylation status
Phospho-proteomic analysis for comprehensive pathway assessment
Time-course experiments to capture signaling dynamics
Research has shown that CA8 overexpression inhibits NGF-induced phosphorylation of ITPR1 and TrkA, as well as ITPR1-mediated cytoplasmic free calcium release . This methodological approach enables detailed mechanistic understanding of CA8's regulatory function.
Based on published research, the following vector systems have proven effective for CA8 overexpression:
AAV8-based vectors:
Key design considerations:
Inclusion of epitope tags (V5) enables discrimination between endogenous and exogenous CA8
Selection of appropriate promoters (CMV for high expression, neuronal-specific promoters for targeted studies)
Codon optimization may improve expression levels
When designing CA8 overexpression experiments, researchers should:
Validate expression using immunoblotting and immunofluorescence
Confirm functional activity through calcium signaling assays
Consider dose-response relationships for physiological relevance
Studies have demonstrated that AAV8-mediated CA8 expression successfully modulates pain responses in preclinical models, making this an optimal approach for neurological applications .
To effectively compare wild-type and mutant CA8 function:
Experimental system selection:
Functional readouts:
ITPR1 phosphorylation status as a direct measure of CA8 activity
Calcium flux measurements following stimulation
Downstream signaling pathway activation
Behavioral outcomes in animal models (e.g., pain sensitivity)
Control considerations:
Empty vector controls
Inactive mutant controls (e.g., CA8-S100P)
Dose-matching between wild-type and mutant constructs
Research has shown that while wild-type human CA8 (CA8 WT) effectively inhibits NGF-induced phosphorylation of ITPR1 and TrkA, the S100P mutation (CA8 MT) fails to produce this inhibitory effect, demonstrating the specificity of CA8's functional domain .
CA8's involvement in pain processing can be investigated through:
Preclinical pain models with CA8 modulation:
Spinal nerve ligation (SNL) model with AAV8-V5-CA8 WT administration
Behavioral assessment of mechanical thresholds using von Frey filaments
Thermal sensitivity testing using Hargreaves apparatus
Longitudinal monitoring to capture therapeutic timeline
Molecular and cellular analysis:
CA8 expression profiling in dorsal root ganglia and spinal cord
Assessment of ITPR1 phosphorylation status in nociceptive neurons
Calcium imaging in primary sensory neurons with CA8 manipulation
Evaluation of neuroinflammatory markers
Data from animal studies demonstrate that AAV8-V5-CA8 WT injection 3 days after SNL resulted in mechanical thresholds returning to baseline by Day 10 and produced analgesia (elevated thresholds above baseline) from Day 24 through Day 38 . In contrast, mutant CA8 (AAV8-V5-CA8 MT) failed to produce similar effects, emphasizing the specificity of wild-type CA8's analgesic properties.
When facing contradictory findings in CA8 research:
Methodological standardization:
Compare expression systems (transient vs. stable, different vector types)
Standardize protein quantification methods (ELISA protocols, antibody selection)
Control for cell type-specific effects (neuronal vs. non-neuronal)
Account for temporal dynamics of calcium signaling
Experimental variables to consider:
Species differences (human CA8 vs. mouse Car8)
Baseline calcium levels in experimental systems
Expression levels of ITPR1 and other CA8 interaction partners
Presence of endogenous CA8 in the model system
Systematic comparative analysis:
Side-by-side testing of wildtype and mutant CA8 variants
Dose-response studies for CA8 expression levels
Multiple readouts of CA8 function (protein interaction, calcium signaling, downstream effects)
When designing resolution experiments, consider that some cell lines show minimal endogenous CA8 expression but high levels of ITPR1 (e.g., NBL cells), making them ideal systems for CA8 functional studies without interference from endogenous protein .
Translating CA8 research to clinical applications faces several challenges:
Delivery challenges:
Vector selection for human translation (AAV8 shows promise in preclinical models)
Targeting specificity to relevant neuronal populations
Achieving therapeutic expression levels without toxicity
Duration of expression for chronic conditions
Safety considerations:
Off-target effects on calcium signaling in non-target tissues
Immune responses to viral vectors or the CA8 protein itself
Long-term consequences of CA8 overexpression
Potential for insertional mutagenesis with integrating vectors
Efficacy assessment:
Development of biomarkers for CA8 activity in humans
Standardized protocols for measuring calcium signaling in accessible tissues
Clinical outcome measures relevant to CA8's physiological effects
Dosing optimization for therapeutic benefit
Researchers pursuing translational applications should consider establishing correlation between CA8 levels in accessible fluids (measured via ELISA) and functional outcomes to facilitate clinical monitoring .
Sample preparation is critical for reliable CA8 analysis:
For serum and plasma:
For cell and tissue lysates:
Lysis in buffers containing protease inhibitors
Avoiding calcium chelators that may affect CA8-ITPR1 interactions
Subcellular fractionation when investigating specific compartments
Protein concentration standardization across samples
For histological analysis:
Fixation optimization (4% paraformaldehyde preferred)
Antigen retrieval methods specific for CA8 epitopes
Blocking endogenous peroxidases and biotin when applicable
Use of appropriate isotype controls
ELISA protocols specifically developed for human CA8 employ antibodies coated on 96-well plates with detection limits typically in the pg/ml range, allowing sensitive quantification across various sample types .
For generating CA8 loss-of-function models:
CRISPR-Cas9 genome editing:
Design of sgRNAs targeting early exons of CA8
Verification of edits by sequencing
Functional validation through calcium signaling assays
Off-target analysis using whole-genome sequencing
RNA interference approaches:
Selection of effective siRNA or shRNA sequences
Stable expression using viral vectors for long-term studies
Titration of knockdown efficiency
Controls for off-target effects
Validation requirements:
Confirmation of CA8 protein reduction (Western blot, ELISA)
mRNA quantification (qRT-PCR)
Assessment of ITPR1 phosphorylation status
Calcium response to ITPR1 agonists
Phenotypic characterization (e.g., pain sensitivity in animal models)
Researchers should be aware that complete CA8 knockout may produce compensatory changes in calcium signaling pathways, potentially complicating interpretation of results. Conditional or inducible systems may help mitigate these confounding effects.
Integrating CA8 data across experimental platforms requires:
Data standardization approaches:
Normalization methods appropriate to each data type
Use of common reference standards
Metadata documentation including experimental conditions
Statistical approaches for cross-platform comparison
Multi-omics integration strategies:
Correlation analysis between CA8 protein levels and transcriptomic data
Pathway analysis incorporating CA8-related signaling components
Network modeling of CA8 interactions
Integration of functional and molecular datasets
Validation strategies:
Key findings validation using orthogonal methods
Replication in independent sample sets
Cross-species validation when appropriate
Functional validation of correlative findings
Researchers should consider developing a CA8-specific database or contributing to existing repositories to facilitate data sharing and integration across studies, enhancing reproducibility and accelerating discovery in this field.
Advanced technologies for investigating CA8's role in calcium signaling include:
Genetically encoded calcium indicators (GECIs):
GCaMP variants for cell-specific calcium imaging
Red-shifted indicators for multiplexing with other fluorescent proteins
FRET-based indicators for ratiometric measurements
Coupling with optogenetic tools for precise stimulation
Super-resolution imaging approaches:
STORM/PALM for nanoscale localization of CA8 and ITPR1
Expansion microscopy for improved spatial resolution
Live-cell super-resolution for dynamic interaction studies
Correlative light and electron microscopy for ultrastructural context
Biosensor development:
FRET-based sensors for CA8-ITPR1 interaction
Conformational sensors for CA8 activation states
Multiplexed sensors for simultaneous pathway monitoring
Single-molecule tracking for diffusion and binding kinetics
These emerging technologies will enable researchers to visualize CA8 function with unprecedented spatial and temporal resolution, providing deeper insights into its regulatory mechanisms in calcium signaling.
Optimization of CA8 gene therapy requires addressing:
Vector engineering advancements:
Capsid modification for enhanced neuronal tropism
Regulated expression systems (inducible promoters)
Reduced immunogenicity through capsid engineering
Improved production methods for clinical-grade vectors
Delivery optimization:
Route selection based on target neuronal populations
Dosing studies to determine minimum effective dose
Timing of intervention (preventative vs. therapeutic)
Co-delivery with adjunct therapies
Efficacy enhancement strategies:
Protein engineering for improved ITPR1 binding
Combination with complementary pain management approaches
Cell-type specific expression to minimize off-target effects
Pharmacogenomic approaches to identify optimal responders
Preclinical data demonstrates that AAV8-V5-CA8 WT administered 3 days after nerve injury restored mechanical thresholds to baseline within 10 days and produced analgesia by significantly raising mechanical thresholds above baseline from Day 24 through Day 38 . This promising timeline suggests potential for clinical translation in neuropathic pain conditions.
Advancing CA8 research through interdisciplinary approaches:
Systems biology integration:
Computational modeling of CA8 in calcium signaling networks
Multi-omics profiling in CA8-modulated systems
Machine learning approaches to identify novel CA8 functions
Population-scale genetic association studies
Translational research opportunities:
Biomarker development for CA8-related disorders
Patient-derived cellular models (iPSCs) for personalized studies
Clinical correlation studies linking CA8 variants to disease phenotypes
Development of non-genetic CA8 modulators (small molecules, peptides)
Evolutionary and comparative studies:
Cross-species analysis of CA8 function and conservation
Evolutionary pressure analysis on CA8 functional domains
Paralog comparison within the carbonic anhydrase family
Ecological and environmental influences on CA8 variation
By integrating approaches from human genetics, neuroscience, biochemistry, and computational biology, researchers can develop a more comprehensive understanding of CA8's roles beyond its established function in calcium regulation.
Carbonic Anhydrase 8 (CA8) is a member of the carbonic anhydrase family, which is a group of zinc metalloenzymes that catalyze the reversible hydration of carbon dioxide. Despite its name, CA8 is unique among carbonic anhydrases because it lacks catalytic activity. This article delves into the background, structure, function, and clinical significance of CA8, particularly focusing on its recombinant form.
CA8 belongs to the alpha-carbonic anhydrase family, which is characterized by a conserved zinc-binding site. However, CA8 is distinct in that it does not possess the typical enzymatic activity associated with other carbonic anhydrases. This is due to the absence of one or more zinc-binding histidine residues, which are crucial for catalytic function .
Despite its lack of enzymatic activity, CA8 plays a significant role in the human body. It is involved in the regulation of intracellular pH and ion transport. CA8 is highly expressed in the cerebellum, where it is believed to play a role in motor coordination and cognitive functions .
Recombinant CA8 is produced using recombinant DNA technology, which involves inserting the CA8 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant form is used in various research applications to study the protein’s structure, function, and potential therapeutic uses .
Mutations in the CA8 gene have been associated with cerebellar ataxia, mental retardation, and dysequilibrium syndrome type 3 (CMARQ3). These conditions are characterized by impaired motor coordination, intellectual disability, and balance issues . Understanding the role of CA8 in these disorders can provide insights into potential therapeutic targets.
Research on CA8 has revealed its widespread expression in various tissues, including the cerebellum, cerebrum, lung, liver, salivary gland, and stomach . This broad expression pattern suggests that CA8 may have multiple functions beyond its role in the cerebellum.
Recombinant CA8 is used in research to investigate its role in different physiological processes and its potential involvement in various diseases. Studies have shown that CA8 is involved in the PKC-gamma calcium signaling pathway, which is important for neuronal function .