Recombinant Pan troglodytes Cardiotrophin-2 (CTF2)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Pan troglodytes Cardiotrophin-2 (CTF2)

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 .

Production and Purification

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 .

Biological Activity and Function

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 .

Applications in Research

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.

Tables

The following tables summarize relevant information discussed in the preceding sections.

Table 1: CTF2 Production Hosts and Characteristics

Host OrganismPurityPost-Translational ModificationsYieldTurnaround Time
E. coli>98%LimitedHighShort
Yeast>90% (ELISA)ModerateHighShort
Insect CellsNot SpecifiedExtensiveVariesVaries
Mammalian CellsNot SpecifiedExtensiveVariesVaries

Table 2: Potential Research Applications of CTF2

ApplicationDescription
Cell Signaling StudiesInvestigating CTF2-mediated pathways and interactions.
Neurological ResearchExploring CTF2's role in neuronal development and survival.
Hematological StudiesExamining CTF2's impact on platelet production.

Product Specs

Form
Lyophilized powder.
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice is specifically requested in advance (additional charges apply).
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
CTF2Cardiotrophin-2; CT-2; Neuropoietin; Np
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
22-203
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Pan troglodytes (Chimpanzee)
Target Names
CTF2
Target Protein Sequence
ASISPAEPI SQAYSLALYM QKNTSALLRT YLQYQGSPLS DPGFSAPELQ LSSLPPATAF FKTWHALDDG EWLSLAQRAF LALTQHLQLV EDDQSDLNPG SPILLAQLGA ARLRAQGLLG NMAAITTALG LPIPPEEDTL GLAAFGASAF ERKCRGYVVT REYGHWTDRA VRDLALLKAK YSA
Uniprot No.

Target Background

Function
Plays a potentially significant role in neuronal precursor development and maturation.
Database Links
Protein Families
IL-6 superfamily
Subcellular Location
Secreted.

Q&A

What is Recombinant Pan troglodytes Cardiotrophin-2 and how does it compare to human Cardiotrophin-1?

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.

What are the recommended storage and handling protocols for Recombinant Pan troglodytes CTF2?

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

What is the recommended reconstitution procedure for lyophilized CTF2?

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

What are the key factors to consider when designing experiments with CTF2?

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:

    • Positive controls (e.g., known cytokines with similar effects)

    • Negative controls (e.g., vehicle-only treatment)

    • Comparative controls (e.g., human CT-1 or other species' cardiotrophin proteins)

  • 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

What is the recommended dose-response assessment methodology for CTF2?

A comprehensive dose-response assessment for CTF2 should include:

  • Concentration range determination:

    • Based on data from related proteins like CT-1, testing a range from 0.1-100 ng/mL would be appropriate

    • Human CT-1 shows activity with an ED₅₀ of 0.25-1.25 ng/mL in relevant bioassays

  • 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

How should researchers design comparative studies between human Cardiotrophin-1 and Pan troglodytes CTF2?

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

How can transgenic models be used to study CTF2 function in vivo?

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

What signaling pathways are activated by CTF2 and how can they be monitored?

Based on knowledge of related cytokines, CTF2 likely activates several signaling pathways:

  • Primary signaling pathways:

    • JAK/STAT pathway (particularly STAT3)

    • MAPK/ERK pathway

    • PI3K/Akt pathway

    • Potentially gp130-dependent signaling, similar to other IL-6 family cytokines

  • 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

How can researchers evaluate the role of CTF2 in cardiac hypertrophy models?

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

What bioassays are most appropriate for measuring CTF2 activity?

Based on established assays for CT-1 and related cytokines:

Bioassay TypeCell SystemReadoutTime FrameSensitivity Range
Growth/DifferentiationTF-1 cellsProliferation (MTT/MTS)48-72 hours0.1-10 ng/mL
HypertrophyNeonatal cardiomyocytesCell size, protein synthesis24-48 hours0.25-5 ng/mL
Signal TransductionHepG2 cellsSTAT3 phosphorylation15-30 minutes0.1-5 ng/mL
Gene InductionEndothelial cellsIL-6 synthesis4-24 hours1-50 ng/mL
Neuronal SurvivalPrimary neuronsCell viability24-72 hours0.5-20 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

  • Validate with receptor blocking or knockdown experiments

What are the recommended protocols for studying CTF2 receptor binding kinetics?

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)

What animal models are most appropriate for in vivo studies with CTF2?

For in vivo CTF2 studies, consider these models:

Animal ModelAdvantagesLimitationsTypical Dose RangeAdministration Routes
MouseGenetic manipulation options, low cost, established cardiac modelsSpecies differences in receptor distribution10-200 μg/kgIP, IV, SC, Intracardiac
RatLarger size for surgical procedures, established cardiac disease modelsFewer genetic tools than mice5-100 μg/kgIP, IV, SC, Intracardiac
RabbitCardiac physiology more similar to humansHigher cost, fewer reagents available1-50 μg/kgIV, SC
PigCardiac anatomy/physiology very similar to humansHigh cost, complex housing requirements0.5-10 μg/kgIV, Intracardiac
Non-human primatesClosest to human physiology/geneticsHighest cost, ethical considerations0.1-5 μg/kgIV, 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

  • Consistency with preliminary in vitro findings

How should researchers address discrepancies between in vitro and in vivo CTF2 activity?

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

What statistical approaches are recommended for analyzing dose-response data for CTF2?

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

How can researchers differentiate between direct and indirect effects of CTF2 in complex biological systems?

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

How can CTF2 research findings in Pan troglodytes models be translated to human applications?

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

What experimental models best simulate pathological conditions where CTF2 may play a role?

Pathological ConditionRecommended ModelsKey ReadoutsRelevance to CTF2
Cardiac HypertrophyPressure overload (TAC), Ang II infusion, Cardiomyocyte stretchHeart weight/body weight ratio, wall thickness, gene expressionLikely induces or responds to hypertrophic stimuli, similar to CT-1
Heart FailureMI model, pressure overload, doxorubicin toxicityEjection fraction, end-diastolic volume, BNP levelsMay have cardioprotective effects like CT-1 in stress conditions
Inflammatory DisordersLPS challenge, sterile inflammation modelsCytokine levels, tissue damage markers, cellular infiltrationMight modulate inflammatory responses via IL-6 family signaling
Metabolic DysfunctionDiet-induced obesity, diabetes modelsGlucose tolerance, insulin sensitivity, tissue metabolismIL-6 family cytokines influence metabolic regulation
Neurological ConditionsNeurotoxic challenges, axonal injury modelsNeuronal survival, axonal regeneration, behavioral testingMay have neurotrophic properties similar to CNTF

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

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.