Recombinant Pan troglodytes Cardiotrophin-2 (CTF2) is a synthetically produced version of the CTF2 protein, derived from chimpanzees (Pan troglodytes). CTF2, also known as cardiotrophin-like cytokine factor 1 (CLCF1), is a cytokine belonging to the IL-6 superfamily . Cytokines are small proteins important in cell signaling, and CTF2 may have a role in neuronal precursor development and increasing platelet count in the presence of splenomegaly . The recombinant form is produced in host cells like Escherichia coli or yeast to generate large quantities of the protein for research and potential therapeutic applications .
Recombinant CTF2 can be expressed and purified from different hosts, including E. coli, yeast, insect, and mammalian cells . E. coli and yeast expression systems often provide the best yields and shorter turnaround times. Expression in insect or mammalian cells can provide post-translational modifications necessary for correct protein folding and activity . CTF2 produced in E. coli is greater than 98% pure, while CTF2 from yeast is greater than 90% pure as measured by ELISA .
The precise biological activity of Pan troglodytes CTF2 is still being investigated, but, generally, CTF2 is associated with:
Neuronal Development: CTF2 may play a role in the development of neuronal precursors .
Platelet Count Regulation: CTF2 increases platelet count in the presence of splenomegaly .
Cytokine Activity: As a member of the IL-6 superfamily, CTF2 likely participates in various cell signaling pathways, influencing cell growth, differentiation, and immune responses .
Recombinant Pan troglodytes CTF2 is primarily used in preclinical research to study its effects on various biological systems. Some potential applications include:
Cell Signaling Studies: Investigating the signaling pathways activated by CTF2 and its interaction with other cytokines and receptors.
Neurological Research: Exploring the role of CTF2 in neuronal development, survival, and regeneration.
Hematological Studies: Examining the impact of CTF2 on platelet production and its potential therapeutic use in thrombocytopenia.
The following tables summarize relevant information discussed in the preceding sections.
| Host Organism | Purity | Post-Translational Modifications | Yield | Turnaround Time |
|---|---|---|---|---|
| E. coli | >98% | Limited | High | Short |
| Yeast | >90% (ELISA) | Moderate | High | Short |
| Insect Cells | Not Specified | Extensive | Varies | Varies |
| Mammalian Cells | Not Specified | Extensive | Varies | Varies |
| Application | Description |
|---|---|
| Cell Signaling Studies | Investigating CTF2-mediated pathways and interactions. |
| Neurological Research | Exploring CTF2's role in neuronal development and survival. |
| Hematological Studies | Examining CTF2's impact on platelet production. |
Recombinant Pan troglodytes Cardiotrophin-2 (CTF2) is a cytokine protein derived from chimpanzee (Pan troglodytes) genetic material and produced through recombinant DNA technology. It belongs to the same cytokine family as human Cardiotrophin-1 (CT-1), which includes interleukin-6 (IL-6), IL-11, leukemia inhibitory factor (LIF), oncostatin M (OSM), and ciliary neurotrophic factor (CNTF) .
While human CT-1 was originally isolated based on its ability to induce cardiac myocyte hypertrophy , CTF2 likely shares similar but distinct biological activities. The amino acid sequence of CTF2 would be expected to show high homology with human CT proteins, but with specific differences that may affect receptor binding affinity and downstream signaling pathways.
Based on established protocols for similar recombinant proteins:
Store lyophilized CTF2 at -20°C to -80°C
Reconstituted protein should be stored at -80°C in single-use aliquots
Avoid repeated freeze-thaw cycles as they may compromise protein activity
Reconstitute in sterile buffer (typically PBS or similar buffer with 0.1% carrier protein)
Working solutions should be prepared fresh and used within 24 hours
When handling, use sterile technique and low-protein binding tubes to minimize loss
Reconstitution should follow these steps:
Allow the lyophilized protein to reach room temperature
Reconstitute using sterile, filtered buffer appropriate for your experimental conditions
Gently mix by swirling or rotating; avoid vortexing to prevent protein denaturation
Allow complete solubilization (approximately 10-30 minutes at room temperature)
Aliquot into sterile microcentrifuge tubes for single use
Flash freeze aliquots in liquid nitrogen before storing at -80°C
When designing experiments with CTF2, researchers should apply principles of effective experimental design to ensure valid and reliable results. Key considerations include:
Identifying research questions and hypotheses: Clearly define what you aim to test regarding CTF2's function, based on existing literature on cardiotrophin proteins
Variables management:
Independent variables: The aspects you will manipulate, such as CTF2 concentration, exposure time, or cell types
Dependent variables: The outcomes you will measure, such as cell hypertrophy, protein expression, or pathway activation
Control for extraneous variables: Factors like temperature, cell passage number, and media composition
Controls selection:
Experimental groups: Define clear experimental and control groups with appropriate sample sizes determined through power analysis
Randomization and blinding: Implement where appropriate to minimize bias, especially in animal studies or when analyzing subjective outcomes
A comprehensive dose-response assessment for CTF2 should include:
Concentration range determination:
Experimental design approach:
Use a logarithmic scale of concentrations (e.g., 0.1, 0.3, 1, 3, 10, 30, 100 ng/mL)
Include technical and biological replicates (minimum n=3 for each)
Include time-course elements (response at different time points)
Analysis methods:
Calculate EC₅₀/ED₅₀ values using nonlinear regression
Determine Hill slope to understand cooperativity
Compare with known cytokines like human CT-1 for reference
Validation steps:
Confirm specificity using receptor antagonists or competitive inhibitors
Verify results across different cell types relevant to CTF2 function
For comparative studies between human CT-1 and Pan troglodytes CTF2:
Experimental approach: Use a true experimental design with controlled variables and randomization
Matched conditions: Ensure identical experimental conditions for both proteins, including:
Same cell lines/primary cells
Identical medium formulations
Parallel processing and analysis
Same detection methods and reagents
Cross-species considerations:
Test on both human and chimpanzee cells when possible
Include species-matched receptor systems
Consider receptor antagonists to confirm specificity
Analysis framework:
Direct statistical comparison of dose-response curves
Analysis of receptor binding kinetics
Comparison of downstream signaling pathways
Assessment of biological outcomes (e.g., hypertrophy, proliferation)
Controls:
Include other IL-6 family cytokines as references
Use species-matched controls where appropriate
Transgenic models offer powerful tools for studying CTF2 function in vivo:
Types of transgenic approaches:
Overexpression models (cardiac-specific or inducible)
Knockout/knockdown models
Humanized models (replacing endogenous CTF with human version)
Reporter models (CTF2 promoter driving reporter gene expression)
Experimental design considerations:
Use appropriate controls, including littermate wild-type animals
Consider tissue-specific or inducible expression systems to avoid developmental effects
Account for genetic background effects by backcrossing to pure strains
Design longitudinal studies to observe temporal effects
Phenotypic analyses:
Cardiac function (echocardiography, pressure-volume loops)
Histological assessment (hypertrophy, fibrosis)
Molecular analyses (transcriptomics, proteomics)
Response to stress conditions (ischemia-reperfusion, pressure overload)
Translational relevance:
Correlation with human disease models
Therapeutic targeting potential
Biomarker development
Based on knowledge of related cytokines, CTF2 likely activates several signaling pathways:
Primary signaling pathways:
Monitoring methods:
Western blotting for phosphorylated signaling proteins
Immunofluorescence for nuclear translocation of transcription factors
Reporter gene assays (e.g., STAT3-responsive luciferase reporters)
Phosphoproteomic analysis for comprehensive pathway mapping
CRISPR-mediated HiBiT-tagging for monitoring phosphorylation and acetylation
Temporal considerations:
Immediate-early responses (minutes to hours)
Secondary responses (hours to days)
Feedback regulation and pathway crosstalk
Inhibitor studies:
Pathway-specific inhibitors to confirm causality
siRNA knockdown of pathway components
Receptor antagonists or neutralizing antibodies
To evaluate CTF2's role in cardiac hypertrophy:
In vitro models:
Primary cardiomyocyte cultures (neonatal or adult)
Cardiomyocyte cell lines
3D cardiac organoids
Assessing hypertrophy:
Cell size measurement (immunofluorescence, flow cytometry)
Protein synthesis (³H-leucine incorporation)
Hypertrophic gene expression (ANP, BNP, β-MHC)
Sarcomere organization (α-actinin staining)
In vivo models:
Pressure overload (transverse aortic constriction)
Volume overload (aortocaval fistula)
Isoproterenol-induced hypertrophy
Post-myocardial infarction remodeling
Mechanistic studies:
CTF2 gain and loss of function
Receptor antagonism
Downstream pathway inhibition
Integration with other hypertrophic stimuli
Based on established assays for CT-1 and related cytokines:
| Bioassay Type | Cell System | Readout | Time Frame | Sensitivity Range |
|---|---|---|---|---|
| Growth/Differentiation | TF-1 cells | Proliferation (MTT/MTS) | 48-72 hours | 0.1-10 ng/mL |
| Hypertrophy | Neonatal cardiomyocytes | Cell size, protein synthesis | 24-48 hours | 0.25-5 ng/mL |
| Signal Transduction | HepG2 cells | STAT3 phosphorylation | 15-30 minutes | 0.1-5 ng/mL |
| Gene Induction | Endothelial cells | IL-6 synthesis | 4-24 hours | 1-50 ng/mL |
| Neuronal Survival | Primary neurons | Cell viability | 24-72 hours | 0.5-20 ng/mL |
When selecting a bioassay:
Choose biologically relevant cell types (cardiac, hepatic, neuronal)
Include appropriate positive controls (e.g., CT-1, LIF)
Test multiple concentrations to establish dose-response relationships
For studying CTF2 receptor binding kinetics:
Preparation of labeled CTF2:
Radioactive labeling (¹²⁵I)
Fluorescent labeling (AlexaFluor dyes)
Biotinylation for streptavidin-based detection
Binding assay methods:
Saturation binding assays (increasing concentrations of labeled CTF2)
Competition binding assays (fixed labeled CTF2 with increasing unlabeled competitor)
Association/dissociation kinetics (time course measurements)
Analysis approaches:
Scatchard plots or nonlinear regression for K₁ determination
Competitive binding curves for IC₅₀ values
Association/dissociation rate constants (kon and koff)
Advanced techniques:
Surface plasmon resonance (SPR)
Biolayer interferometry (BLI)
Microscale thermophoresis (MST)
Fluorescence resonance energy transfer (FRET)
For in vivo CTF2 studies, consider these models:
| Animal Model | Advantages | Limitations | Typical Dose Range | Administration Routes |
|---|---|---|---|---|
| Mouse | Genetic manipulation options, low cost, established cardiac models | Species differences in receptor distribution | 10-200 μg/kg | IP, IV, SC, Intracardiac |
| Rat | Larger size for surgical procedures, established cardiac disease models | Fewer genetic tools than mice | 5-100 μg/kg | IP, IV, SC, Intracardiac |
| Rabbit | Cardiac physiology more similar to humans | Higher cost, fewer reagents available | 1-50 μg/kg | IV, SC |
| Pig | Cardiac anatomy/physiology very similar to humans | High cost, complex housing requirements | 0.5-10 μg/kg | IV, Intracardiac |
| Non-human primates | Closest to human physiology/genetics | Highest cost, ethical considerations | 0.1-5 μg/kg | IV, SC |
Key considerations for animal model selection:
Research question specificity (cardiac, hepatic, neurological effects)
Species differences in receptor expression and signaling
Availability of supporting reagents (antibodies, assays)
Ethical considerations and regulatory requirements
When confronting discrepancies between in vitro and in vivo CTF2 activity:
Systematic evaluation:
Document all differences systematically
Consider dose-response relationships in both systems
Evaluate temporal dynamics (acute vs. chronic effects)
Assess endpoint relevance and measurement techniques
Biological explanations:
Receptor expression differences between cell culture and intact tissues
Presence of binding proteins or antagonists in vivo
Compensatory mechanisms present in vivo but absent in vitro
Pharmacokinetic considerations (distribution, metabolism, clearance)
Complex intercellular communication in intact systems
Technical considerations:
Protein stability differences between systems
Delivery method limitations in vivo
Detection sensitivity variations
Experimental conditions (temperature, pH, oxygen levels)
Resolution approaches:
Develop more physiologically relevant in vitro systems (3D cultures, co-cultures)
Use ex vivo approaches (isolated organ preparations, tissue slices)
Consider intermediate complexity models (organoids)
Refine in vivo delivery methods or dosing regimens
Employ genetic approaches to validate pharmacological findings
For analyzing CTF2 dose-response data:
Preliminary data analysis:
Assess normality (Shapiro-Wilk test)
Identify outliers (Grubbs' test)
Transform data if needed (log transformation often appropriate for biological responses)
Dose-response modeling:
Four-parameter logistic regression (preferred for sigmoidal responses)
Three-parameter models when appropriate (fixed Hill slope or bottom/top)
Specialized models for bell-shaped or biphasic responses
Goodness-of-fit assessment (R², residual analysis)
Parameter extraction and comparison:
EC₅₀/IC₅₀ determination with confidence intervals
Maximum effect (Emax) assessment
Hill slope evaluation for mechanistic insights
Area under the curve analysis for integrated responses
Advanced statistical approaches:
Global fitting for comparing multiple dose-response curves
Extra sum-of-squares F test for model comparison
Bootstrapping for robust confidence interval estimation
Mixed-effects models for handling repeated measures
Visualization:
Semi-logarithmic plotting (log concentration vs. response)
Include individual data points with curve fit
Clearly indicate error bars (SEM or 95% CI)
Use consistent scales when comparing multiple conditions
To differentiate between direct and indirect effects of CTF2:
Experimental strategies:
Time-course studies (direct effects typically occur earlier)
Protein synthesis inhibition (cycloheximide) to block secondary effects
Receptor antagonism or knockdown in specific cell types
Cell-specific genetic deletion in complex systems
Ex vivo studies with isolated cell populations
Conditioned media experiments to identify secreted mediators
Signaling pathway analysis:
Immediate receptor-proximal events (typically direct)
Secondary messenger activation patterns
Transcriptional profiling with temporal resolution
Phosphoproteomic analysis at multiple time points
Pathway inhibitor panels with varying specificity
Systems biology approaches:
Network analysis of responding genes/proteins
Computational modeling of direct and indirect interactions
Bayesian network inference from multi-parameter data
Integration of transcriptomic and proteomic datasets
Validation approaches:
Independent methodologies confirmation
In vitro reconstitution with defined components
Cross-validation in multiple model systems
Correlation with known direct targets of related cytokines
Translating CTF2 research from Pan troglodytes to human applications requires:
Comparative analysis:
Sequence homology assessment between chimpanzee CTF2 and human orthologs
Receptor binding studies in both species' cell types
Signaling pathway conservation analysis
Functional conservation testing in parallel assays
Translational models:
Humanized mouse models expressing human receptors
Human cell and tissue systems (iPSC-derived cells, organoids)
Ex vivo human tissue studies where ethically possible
Computational prediction of cross-species differences
Biomarker development:
Identify conserved biomarkers of CTF2 activity
Develop assays applicable to human samples
Correlate markers with functional outcomes
Validate in human pathological samples when available
Therapeutic considerations:
Species-specific differences in pharmacokinetics
Immunogenicity risk assessment for non-human proteins
Humanization strategies for therapeutic development
Target validation in human systems
For each model:
Consider both acute and chronic experimental designs
Include genetic manipulation of CTF2 or its receptors
Employ pharmacological interventions (recombinant protein, antagonists)
Measure both molecular and functional outcomes
Include time-course studies to capture dynamic responses