Recombinant Human Transthyretin (TTR) (Active) is a laboratory-produced, biologically functional form of the human transthyretin protein, optimized for research and therapeutic development. TTR is a 55 kDa homotetrameric serum protein primarily involved in transporting thyroid hormones (T4/T3) and retinol (via binding to retinol-binding protein, RBP) . Its recombinant form is engineered to retain native structural stability and ligand-binding capabilities, enabling studies on amyloidosis mechanisms, neuroprotection, and drug discovery .
Key functional roles include:
Amyloidosis modulation: Stabilizes tetrameric TTR to inhibit pathological fibril formation in familial amyloidotic polyneuropathy (FAP) and cardiomyopathy .
Neuroprotection: Binds amyloid-β (Aβ) in Alzheimer’s disease (AD) and α-synuclein in Parkinson’s disease (PD), suppressing toxic aggregation .
Thyroid hormone transport: Maintains systemic thyroxine levels, with implications for metabolic regulation .
Expression Hosts: HEK293 cells (most common), E. coli (limited due to folding challenges) .
Purification: Affinity chromatography (His-tag), SEC for oligomer separation, >90% purity confirmed by SDS-PAGE .
Kinetic Turbidimetry: Measures fibril formation rates (absorbance at 340 nm) with inhibitors like diflunisal or tafamidis. Parameters include IC₅₀ (3–15 μM) and RA(%) (60–100% inhibition) .
Acid-Induced Fibrillogenesis: pH 4.4 incubation triggers TTR aggregation, monitored via Thioflavin T fluorescence .
α-Synuclein Cleavage: oTTR (not tTTR) cleaves free α-synuclein, preventing amyloid formation (IC₅₀ = 6 nM) .
Aβ Interaction: TTR degrades Aβ via conformation-selective proteolysis, reducing neurotoxicity in AD models .
Alzheimer’s Disease: TTR binds Aβ in CSF, reducing plaque deposition by 60–75% in transgenic mouse models .
Parkinson’s Disease: Proteolytic cleavage of α-synuclein by oTTR delays fibril formation, with IC₅₀ values as low as 0.147 μM .
Stroke Recovery: High serum TTR correlates with improved outcomes in cerebral infarction patients .
| TTR Variant | Tetramer Stability | Amyloidogenicity |
|---|---|---|
| Wild-Type (WT) | High | Low |
| Mutant (e.g., V30M) | Reduced | High |
| Drug-Stabilized (e.g., tafamidis) | Enhanced | Inhibited |
Data sourced from in vitro assays and transgenic models .
TTR Stabilizers: Diflunisal, tafamidis, and luteolin enhance tetramer stability, delaying amyloidosis progression .
Redox Modulators: N-acetylcysteine (NAC) suppresses oxidative misfolding of TTR-V30M in plasma .
Oligomer Dynamics: oTTR’s role in proteolysis requires further characterization for PD/AD therapeutic targeting .
Delivery Systems: Enhancing blood-brain barrier penetration for CNS amyloidosis treatment .
Standardization: Batch-to-batch variability in oTTR content (4–7%) impacts assay reproducibility .
DAZAP2 (DAZ-associated protein 2) is a 108 amino acid proline-rich protein that plays crucial roles in multiple cellular processes including spermatogenesis, RNA splicing, transcription regulation, and cell signaling. Its significance has expanded considerably with recent discoveries of its function as a pan-coronavirus restriction factor. DAZAP2 is primarily located in the nucleus, though it was previously thought to be cytoplasmic, and is encoded by a gene on human chromosome 12. The protein's interactions with other key proteins such as Sox-6, DAZL, and DAZ (deleted in azoospermia) highlight its importance in both reproductive biology and broader cellular functions. Recent research has revealed its unexpected role in innate immunity against coronaviruses, making it a target of significant interest for virologists and immunologists .
The most commonly used DAZAP2 antibody in research is the mouse monoclonal IgG1 kappa light chain antibody (such as the G-4 clone). These antibodies are available in both non-conjugated forms and various conjugated formats including:
Agarose-conjugated for immunoprecipitation
Horseradish peroxidase (HRP)-conjugated for enhanced chemiluminescence detection
Fluorescent conjugates including phycoerythrin (PE), fluorescein isothiocyanate (FITC)
Multiple Alexa Fluor® conjugates for fluorescence microscopy and flow cytometry
These diverse formats allow researchers to select the appropriate antibody configuration based on their specific experimental requirements and detection methods .
The specificity of DAZAP2 antibodies is typically confirmed through multiple validation techniques to ensure cross-species reactivity. High-quality DAZAP2 antibodies, such as the G-4 clone, are verified to detect DAZAP2 protein from mouse, rat, and human origins. Validation methods include:
Western blotting with lysates from multiple species
Immunoprecipitation followed by mass spectrometry
Immunofluorescence with parallel siRNA knockdown controls
Cross-validation with multiple antibodies targeting different epitopes
Testing on DAZAP2 knockout cell lines (particularly important in coronavirus research)
Researchers should review validation data and consider using DAZAP2 knockout models as negative controls, especially when studying novel functions of DAZAP2 in virus restriction .
DAZAP2 antibodies are versatile tools employed in multiple research techniques:
Western Blotting (WB): For detection and quantification of DAZAP2 protein expression levels in cell or tissue lysates. Typically run on 12-15% gels due to DAZAP2's relatively small size (108 amino acids).
Immunoprecipitation (IP): For isolation of DAZAP2 and its interacting partners to study protein-protein interactions, particularly with DAZ, Sox-6, and DAZL.
Immunofluorescence (IF): For visualization of DAZAP2's subcellular localization. Recent findings show primarily nuclear localization, contrary to earlier cytoplasmic reports, particularly relevant for coronavirus research.
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative measurement of DAZAP2 levels in biological samples.
Chromatin Immunoprecipitation (ChIP): For investigating DAZAP2's potential role in transcriptional regulation.
Flow Cytometry: Using fluorophore-conjugated antibodies to analyze DAZAP2 expression in different cell populations .
Recent breakthrough research has identified DAZAP2 as a pan-coronavirus restriction factor, opening new applications for DAZAP2 antibodies in virology research:
Immunofluorescence co-localization studies: To determine if DAZAP2 co-localizes with viral proteins or cellular structures involved in viral entry or replication. Research shows DAZAP2 does not co-localize with early endosome marker EEA1 or endolysosome marker LAMP1, suggesting its antiviral mechanism is indirect.
Proximity ligation assays: To detect potential interactions between DAZAP2 and viral components.
Co-immunoprecipitation: To identify host factors that interact with DAZAP2 during viral infection, potentially revealing its mechanism of action.
Immunoblotting in viral infection time-course studies: To monitor DAZAP2 expression and potential post-translational modifications during infection.
Quantitative analysis in wild-type versus DAZAP2 knockout cells: To measure differences in viral entry, replication, and assembly using appropriate viral markers .
When employing DAZAP2 antibodies in coronavirus research, several methodological adaptations are necessary:
Fixation protocols: Use 4% paraformaldehyde fixation for 15-20 minutes for optimal preservation of both DAZAP2 and viral antigens.
Nuclear protein extraction: Since DAZAP2 primarily localizes to the nucleus, nuclear extraction protocols should be optimized for efficient isolation.
Dual staining procedures: When co-staining for DAZAP2 and viral proteins, careful antibody selection is required to avoid cross-reactivity. Sequential staining may be preferable to simultaneous staining.
Blocking optimization: In cells with high DAZAP2 expression, increased blocking (5-10% normal serum) may be necessary to reduce background.
Controls: Include DAZAP2 knockout cells as essential negative controls in all experiments.
Biosafety considerations: All experiments with infectious coronaviruses must be conducted under appropriate biosafety levels (BSL-3 for SARS-CoV-2) .
Designing rigorous experiments to study DAZAP2's antiviral activity requires careful planning:
Cell model selection: Use physiologically relevant models such as human airway epithelial cells, A549-ACE2 cells for SARS-CoV-2, or appropriate models for other coronaviruses.
DAZAP2 manipulation approaches:
CRISPR/Cas9 knockout (complete gene deletion)
siRNA/shRNA knockdown (transient/stable reduction)
Overexpression systems (for gain-of-function studies)
Mutant DAZAP2 expression (to identify functional domains)
Viral assays:
Pseudotyped virus systems for entry studies
Replicon systems for replication analysis
Authentic virus infection for complete life cycle assessment
Split NanoLuc luciferase-based assays for quantifying cell-cell fusion
Time-course analysis: Examine multiple timepoints post-infection to distinguish effects on early versus late viral life cycle stages.
Complementary approaches: Combine immunofluorescence, qRT-PCR, Western blotting, and functional assays to build a comprehensive understanding of DAZAP2's mechanisms .
Rigorous immunofluorescence studies with DAZAP2 antibodies require comprehensive controls:
Primary antibody controls:
DAZAP2 knockout cells as negative controls
Multiple DAZAP2 antibodies targeting different epitopes
Isotype control antibodies to assess non-specific binding
Secondary antibody controls:
Secondary-only controls (no primary antibody)
Cross-adsorbed secondaries to minimize species cross-reactivity
Expression controls:
siRNA knockdown samples showing reduced signal
Overexpression samples showing increased signal
Subcellular localization controls:
Nuclear markers (e.g., DAPI) to confirm DAZAP2's nuclear localization
Co-staining with organelle markers (e.g., EEA1, LAMP1) to assess potential co-localization
Specificity validation:
To properly analyze DAZAP2's role in inhibiting viral fusion, researchers should consider these advanced methodological approaches:
Quantitative cell-cell fusion assays: Utilize the split NanoLuc luciferase system where:
Acceptor cells express the LgBit fragment
Donor cells express the HiBit fragment and viral spike protein
Upon fusion, complementation forms functional luciferase for quantification
Confocal microscopy analysis:
Measure syncytia formation area
Count syncytial nuclei per field
Quantify fusion kinetics through time-lapse imaging
Membrane dynamics assessment:
Lipid mixing assays with labeled membranes
Content mixing assays to confirm full fusion
Fusion mechanism dissection:
Compare endolysosomal fusion (pH-dependent) versus plasma membrane fusion (TMPRSS2-dependent)
Analyze the impact of pH modulation using bafilomycin A1 or ammonium chloride
Molecular interaction studies:
For effective co-immunoprecipitation (co-IP) experiments with DAZAP2 antibodies:
Antibody selection and immobilization:
Use agarose-conjugated DAZAP2 antibodies for direct IP
For unconjugated antibodies, pre-bind to Protein A/G beads
Consider site-specific biotinylated antibodies with streptavidin beads for oriented immobilization
Lysis buffer optimization:
Use gentle, non-denaturing conditions (e.g., 1% NP-40 or 0.5% CHAPS)
Include protease/phosphatase inhibitors
For nuclear proteins, include benzonase or other nucleases
Test multiple salt concentrations (150-300mM) to optimize specificity
Cross-linking considerations:
For transient interactions, consider reversible cross-linkers (DSP)
For nuclear interactions, consider formaldehyde cross-linking
Washing stringency optimization:
Balance between preserving interactions and reducing background
Consider graduated washing with increasing salt concentrations
Validation approaches:
DAZAP2 has been implicated in multiple myeloma pathogenesis, making it an important research target:
Expression analysis methodology:
Compare DAZAP2 protein levels in multiple myeloma versus normal plasma cells using quantitative Western blot
Perform immunohistochemistry on bone marrow biopsies with appropriate controls
Correlate DAZAP2 expression with clinical outcomes and disease progression
Functional assays:
Use DAZAP2 antibodies to study its interactions with key signaling pathways in myeloma
Investigate effects of DAZAP2 knockdown/overexpression on myeloma cell proliferation, apoptosis, and drug resistance
Explore DAZAP2's potential role in the bone marrow microenvironment
Post-translational modification studies:
Use modification-specific antibodies to detect changes in DAZAP2 phosphorylation, ubiquitination, or other modifications in disease states
Correlate modifications with altered function or localization
Therapeutic implications:
DAZAP2's interaction with DAZ and DAZL proteins makes it relevant to male fertility research:
Tissue-specific expression analysis:
Compare DAZAP2 expression in testicular biopsies from fertile versus infertile men
Characterize DAZAP2 expression during different stages of spermatogenesis
Co-localize DAZAP2 with DAZ/DAZL in testicular cells
Protein-protein interaction studies:
Use co-immunoprecipitation with DAZAP2 antibodies to pull down DAZ/DAZL complexes
Map interaction domains through mutational analysis
Investigate how these interactions affect RNA processing
Functional consequences:
Examine DAZAP2 knockout models for spermatogenesis defects
Analyze DAZAP2 mutations or polymorphisms in infertile populations
Study the impact of DAZAP2 variations on DAZ/DAZL function
RNA-related functions:
To robustly demonstrate DAZAP2's role as a pan-coronavirus restriction factor, researchers should employ:
Virus diversity testing:
Challenge DAZAP2-deficient versus control cells with coronaviruses from different genera
Test against alpha-coronaviruses (e.g., HCoV-229E, HCoV-NL63)
Test against beta-coronaviruses (e.g., SARS-CoV-2, SARS-CoV, MERS-CoV)
Test against gamma and delta coronaviruses when possible
Mechanistic dissection:
Viral entry assays using pseudotyped particles
Replicon systems to isolate replication effects
Time-of-addition experiments with DAZAP2 overexpression
Split entry pathways using specific inhibitors (e.g., cathepsin inhibitors for endosomal entry, TMPRSS2 inhibitors for surface entry)
In vivo validation:
Mouse models with DAZAP2 knockout
Primary cell cultures including human airway epithelial cells
Quantitative viral load measurements in different tissues
Comparative genomics:
When encountering conflicting data regarding DAZAP2's subcellular localization:
Technical considerations:
Evaluate fixation methods (paraformaldehyde versus methanol can yield different results)
Compare antibody clones targeting different DAZAP2 epitopes
Assess specificity controls (knockout validation, peptide competition)
Consider cell type-specific differences in localization
Biological factors:
Investigate potential relocalization during viral infection
Examine cell cycle-dependent localization changes
Assess shuttling between nucleus and cytoplasm
Check for alternatively spliced isoforms with different localization patterns
Resolution approaches:
Perform fractionation studies with biochemical verification
Use live-cell imaging with fluorescently-tagged DAZAP2
Employ super-resolution microscopy for precise localization
Validate with orthogonal techniques (e.g., electron microscopy)
Reconciliation with functional data:
To determine if DAZAP2's antiviral activity depends on innate immune signaling:
Genetic approaches:
Create double knockout cell lines lacking both DAZAP2 and key innate immune components:
DAZAP2/STAT1 double knockout
DAZAP2/MAVS double knockout
DAZAP2/IRF3 double knockout
Compare virus replication in these lines versus single knockouts
Signaling pathway analysis:
Measure type I interferon production in DAZAP2-sufficient versus deficient cells
Analyze ISG (interferon-stimulated gene) induction
Examine NF-κB activation status
Monitor IRF3 phosphorylation and nuclear translocation
Rescue experiments:
Attempt to rescue the DAZAP2 knockout phenotype with exogenous interferon
Test if DAZAP2's antiviral effect persists in cells unable to respond to interferon
Transcriptional profiling:
Compare transcriptomes of wild-type versus DAZAP2-deficient cells before and after infection
Analyze changes in innate immune gene expression
Look for unique gene signatures that might explain DAZAP2's mechanism
Temporal considerations:
When encountering non-specific binding with DAZAP2 antibodies:
Blocking optimization:
Test different blocking agents (BSA, milk, normal serum, commercial blockers)
Increase blocking time (1-2 hours) and concentration (3-5%)
Consider dual blocking with different agents sequentially
Antibody dilution optimization:
Perform titration series to identify optimal concentration
For Western blots, typically start with 1:500-1:2000 dilutions
For IF, typically start with 1:100-1:500 dilutions
Washing modifications:
Increase washing stringency with higher detergent concentrations
Extend washing times and increase the number of washes
Consider adding low salt (50-100mM NaCl) to washing buffers
Sample preparation refinement:
Ensure complete protein denaturation for Western blots
Optimize fixation protocols for immunofluorescence
Consider antigen retrieval methods for formalin-fixed samples
Validation approaches:
For accurate quantification of DAZAP2 expression during infection:
Western blot quantification:
Use internal loading controls (β-actin, GAPDH, or preferably total protein stains)
Apply appropriate normalization methods
Employ digital image analysis software with linear dynamic range
Run biological replicates (n≥3) for statistical analysis
qRT-PCR measurement:
Select stable reference genes verified under infection conditions
Use the 2^(-ΔΔCt) method with appropriate controls
Include no-RT controls and standard curves
Validate primers for specificity and efficiency
Immunofluorescence quantification:
Use consistent exposure settings across all samples
Perform nuclear versus cytoplasmic intensity measurements
Apply automated unbiased image analysis algorithms
Analyze sufficient cell numbers for statistical power
Flow cytometry analysis:
Include fluorescence-minus-one (FMO) controls
Set gates based on negative controls
Measure median fluorescence intensity rather than percent positive
Analyze sufficient events (>10,000) per sample
Statistical considerations:
To properly analyze DAZAP2's dual inhibitory mechanisms (entry inhibition and replication suppression):
Dissection of entry versus replication effects:
Use pseudotyped viruses containing coronavirus spike but non-coronavirus genome to isolate entry effects
Employ replicon systems lacking structural proteins to isolate replication effects
Perform time-of-addition experiments with DAZAP2 expression constructs
Quantitative fusion assays:
Use split reporter systems (e.g., NanoLuc complementation assay)
Compare results in the following experimental conditions:
| Condition | Entry Pathway | Expected Result in DAZAP2-KO |
|---|---|---|
| No treatment | Both pathways | Enhanced fusion |
| Cathepsin inhibitors | Surface only | Enhanced fusion |
| TMPRSS2 inhibitors | Endosomal only | Enhanced fusion |
| Both inhibitors | Neither pathway | No fusion |
Replication analysis techniques:
Quantify genomic RNA via qRT-PCR targeting NSP genes
Measure primary translation using replicons with reporter genes
Use luciferase-based replicon systems for kinetic analysis
The table below outlines expected results:
| Measurement | Timepoint | Control Cells | DAZAP2-KO Cells |
|---|---|---|---|
| Genomic RNA | 2-4h | Low (baseline) | Low (baseline) |
| Genomic RNA | 8-24h | Moderate | High |
| Primary translation | 2-4h | Detectable | Similar to control |
| Replication | 8-24h | Moderate | High |
Mechanistic dissection:
When analyzing DAZAP2 knockout effects in coronavirus models, these statistical approaches are recommended:
In vitro experimental analysis:
For continuous variables (viral titers, reporter activity):
Unpaired t-tests for single timepoint, two-group comparisons
ANOVA with post-hoc tests for multi-group comparisons
Mixed-effects models for time-course experiments
For categorical outcomes (percent infected cells):
Chi-square or Fisher's exact tests
Logistic regression for adjusted analyses
In vivo experimental analysis:
Survival data:
Kaplan-Meier curves with log-rank tests
Cox proportional hazards models for covariate adjustments
Viral load data:
Area-under-curve (AUC) analysis for time-course data
Repeated measures ANOVA or mixed-effects models
Non-parametric alternatives if assumptions aren't met
Sample size considerations:
Power analysis based on preliminary data
Consider effect sizes observed in similar studies
Typically n=8-12 animals per group for in vivo studies
Minimum n=3 independent biological replicates for in vitro work
Reporting standards:
To investigate DAZAP2's therapeutic potential:
Druggable pathway identification:
Perform transcriptomic and proteomic analyses to identify DAZAP2-regulated pathways
Screen for small molecules that enhance DAZAP2 expression or activity
Investigate drugs that target downstream effectors in DAZAP2 pathways
Peptide-based approaches:
Identify minimal functional domains of DAZAP2 with antiviral activity
Design cell-penetrating peptides mimicking these domains
Test peptide efficacy in cellular and animal models
Gene therapy considerations:
Develop DAZAP2 expression vectors for targeted delivery
Optimize expression levels to enhance antiviral activity without toxicity
Test delivery methods (viral vectors, lipid nanoparticles) in animal models
Combination therapy strategies:
Assess synergy between DAZAP2-enhancing compounds and direct-acting antivirals
Evaluate potential in preventing resistance emergence
Test effectiveness against multiple coronavirus strains
Biomarker development:
Critical unresolved questions about DAZAP2's antiviral mechanisms include:
Nuclear-cytoplasmic disconnect:
How does a primarily nuclear protein inhibit cytoplasmic viral entry and replication?
What transcriptional targets of DAZAP2 mediate its antiviral effects?
Is there a small cytoplasmic pool of DAZAP2 with direct antiviral activity?
Evolutionary considerations:
Is DAZAP2's antiviral function conserved across species?
Do coronaviruses encode antagonists that counteract DAZAP2 restriction?
Has DAZAP2 undergone positive selection in response to coronavirus pressure?
Specificity questions:
Why is DAZAP2 effective against coronaviruses but not necessarily other virus families?
What unique features of coronavirus entry or replication are targeted?
Does DAZAP2 recognize specific viral components or patterns?
Regulatory aspects:
What signals regulate DAZAP2 expression during infection?
How is DAZAP2 activity modulated by post-translational modifications?
Does DAZAP2 function as part of a larger restriction complex?
Translational gaps: