CTF1 Human, His

Cardiotrophin-1 Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Core Structure

CTF1 Human, His is a 212-amino-acid protein comprising:

  • 200 residues of native human CT-1 (excluding the His tag) .

  • 12 additional residues forming the N-terminal His tag (MRGSHHHHHHGS) .

  • Molecular mass: ~22.5 kDa , slightly larger than the non-tagged form (21.2 kDa) .

ParameterValueSource
Expression HostEscherichia coli
Purification MethodMetal affinity chromatography (via His tag)
Purity>90% (SDS-PAGE)

Formulation

The protein is lyophilized in 0.05M acetate buffer (pH 4) . For reconstitution, a 0.1M acetate buffer (pH 4) is recommended to achieve a stock concentration of ~0.5 mg/ml. Higher concentrations may reduce solubility .

ParameterSpecificationSource
Storage ConditionLyophilized: -20°C; Reconstituted: 4°C (up to 2 weeks)
Freeze-Thaw CyclesAvoid repeated cycles to preserve activity

Key Functions

  • Cardiac Hypertrophy: Induces hypertrophic responses in cardiac myocytes via gp130/LIFR receptor activation and JAK/STAT signaling .

  • Neuroprotection: Reduces motoneuron death in mice during embryonic and postnatal development .

  • Autophagy Induction: Stimulates autophagy in fibroblasts via STAT3 phosphorylation and AMPK activation .

MechanismPathway/Sigaling ComponentBiological OutcomeSource
STAT3 Phosphorylationgp130/LIFR → JAK/STAT → Nuclear translocationAutophagy gene upregulation (Atg3, Atg7, Atg8)
AMPK ActivationCalmodulin-dependent kinase II → AMPKOxidative metabolism enhancement

Therapeutic Potential

Disease/ConditionMechanism of ActionEvidence Source
Heart FailureReduces hepatocellular damage; elevates heat shock proteins (HSPs)
Insulin ResistanceSNP rs8046707 (CTF1 locus) linked to reduced insulin sensitivity
CancerPromotes stromal autophagy; modulates tumor microenvironment

Genetic Insights

  • CTF1 Polymorphisms:

    • rs8046707 (minor allele): Associated with lower insulin sensitivity (HOMA-IR ↑, ISI OGTT ↓) in non-diabetic populations .

    • rs1458201: Nominally linked to reduced visceral adipose tissue (VAT) .

Table 1: Production and Biochemical Properties

ParameterValueSource
Amino Acid SequenceMRGSHHHHHHGS... (His tag) + native CT-1 (200 residues)
Biological ActivityED₅₀ <1.0 ng/ml (TF-1 cell proliferation assay)
Purity>90% (SDS-PAGE; SEC-MALS verified)

Table 2: CTF1 in Disease Pathology

DiseaseRole of CTF1Source
CardiomyopathyElevated levels correlate with dilated cardiomyopathy and arrhythmia
AgingCTF1 deficiency → reduced arterial stiffness; increased lifespan in mice

Product Specs

Introduction
Cardiotrophin 1 (CT-1), a member of the interleukin-6 superfamily, is a 201 amino acid protein known for its ability to induce hypertrophy in cardiac myocytes. Found in both cardiac myocytes and nonmyocytes, CT-1 mRNA is also abundant in adult human lungs and fetal and adult airway smooth muscle cells. CT-1 activates gp130 dependent signaling and the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, transducing hypertrophic and cytoprotective signals in cardiac myocytes. Furthermore, CT-1 exhibits neurotrophic properties, with deficiencies leading to increased motoneuron cell death in mice spinal cord and brainstem nuclei during early development. Acting as a hepatocyte survival factor, CT-1 can reduce hepatocellular damage in acute liver injury animal models. Hypoxic stimulation increases CT-1 expression, offering protection to cardiac cells when administered before simulated ischemia or during reoxygenation. This protective effect extends to inducing the expression of heat shock proteins (hsps) in cardiac cells. CT-1 also elevates ventricular expression of ANP, brain natriuretic peptide (BNP), and angiotensinogen mRNA. Notably, elevated Cardiophin 1 levels are observed in patients with heart failure, dilatative cardiomyopathy, mitral regurgitation, angina, and after acute myocardial infarction.
Description
Produced in E. coli, the Cardiotrophin His-Tagged Fusion Protein Human is a 22.5 kDa protein. It consists of 200 amino acid residues from human Cardiotrophin and a 12 amino acid residue His Tag (HHHHHH).
Formulation
CTF1 was lyophilized from a 0.5 mg/ml solution in 0.05M Acetate buffer with a pH of 4. The solution was filtered through a 0.4 μm filter before lyophilization.
Solubility
To prepare a working stock solution of approximately 0.5 mg/ml, add 0.1M Acetate buffer (pH 4) to the lyophilized pellet and allow it to dissolve completely. For use at a higher pH, dilute the stock solution to 10 µg/ml with the appropriate buffer. The solubility of this antigen is limited at higher concentrations. This product is not sterile; filter it through a sterile filter before use in cell cultures.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited time, showing no changes for up to two weeks.
Purity
SDS-PAGE analysis indicates that the purity of CTF1 Human Recombinant is greater than 90%.
Synonyms
CTF1, CT1, CT-1, Cardiophin 1.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GSSRREGSLE DPQTDSSVSL LPHLEAKIRQ THSLAHLLTK YAEQLLQEYV QLQGDPFGLPSFSPPRLPVA GLSAPAPSHA GLPVHERLRL DAAALAALPP LLDAVCRRQA ELNPRAPRLL RRLEDAARQA RALGAAVEAL LAALGAANRG PRAEPPAATA SAASATGVFP AKVLGLRVCG LYREWLSRTE GDLGQLLPGG SA.

Q&A

What is CTF1 and what are its primary biological functions?

CTF1 (Cardiotrophin 1) is a secreted cytokine that belongs to the IL-6 family of proteins. Its primary biological function includes inducing cardiac myocyte hypertrophy in vitro through binding and activation of the ILST/gp130 receptor complex. CTF1 plays significant roles in cardiac development and function, with research demonstrating its involvement in both physiological and pathological cardiac conditions. The protein is encoded by the CTF1 gene located on chromosome 16 and exists in at least two isoforms resulting from alternative transcript variants . As a member of the cytokine signaling pathway in the immune system, CTF1 participates in multiple cellular processes beyond cardiac tissue, although cardiac effects remain its most studied function .

How is CTF1 structured genetically and what are its key molecular characteristics?

The CTF1 gene (HGNC: 2499, NCBI Gene: 1489) is a protein-coding gene located on chromosome 16. It encodes a secreted cytokine with characteristic structural features of the IL-6 family. The gene produces at least two transcript variants encoding different isoforms of the protein. At the molecular level, CTF1 contains specific domains that enable receptor binding and downstream signaling capabilities . The protein demonstrates cytokine activity and leukemia inhibitory factor receptor binding capabilities according to Gene Ontology annotations. CTF1's molecular structure facilitates its interaction with the ILST/gp130 receptor complex, enabling the activation of downstream signaling pathways associated with cardiac hypertrophy and other cellular responses .

What are the common applications of His-tagged CTF1 in basic research?

His-tagged CTF1 provides researchers with a purification-optimized version of the protein for numerous experimental applications. The addition of a histidine tag allows for efficient single-step purification using immobilized metal affinity chromatography (IMAC), typically with Ni-NTA resins. This purified protein can be utilized in receptor binding assays to study the interaction with ILST/gp130 receptors, in vitro cardiac myocyte hypertrophy models, and protein-protein interaction studies. The tag also facilitates detection in Western blotting and immunoprecipitation experiments without requiring specific CTF1 antibodies, as anti-His antibodies can be used instead. For cellular studies, recombinant His-tagged CTF1 enables dose-response experiments to determine concentration-dependent effects on signaling pathways associated with cardiac hypertrophy and immune system modulation .

How does CTF1 function as a chromatin domain boundary at telomeres?

CTF1 serves as a chromatin domain boundary element at human telomeres through its dual DNA and histone-binding capabilities. Research has demonstrated that CTF1 can protect genes from telomeric silencing when its DNA-binding sites are strategically positioned between the target gene and telomeric extremity, effectively creating a barrier that prevents the propagation of silencing chromatin modifications . This boundary function is mediated through CTF1's ability to recruit the histone variant H2A.Z and restore high histone acetylation levels to the insulated telomeric transgene. Mechanistically, CTF1 disrupts the spread of hypoacetylated histone structure that typically emanates from telomeres . This function appears specific to creating a boundary, as CTF1 does not affect genes immediately adjacent to telomeres, suggesting a positional requirement for its insulator activity .

What methodological approaches can resolve conflicts in CTF1 functional studies?

When encountering conflicting data in CTF1 functional studies, researchers should implement a multi-faceted approach:

  • Protein source verification: Compare recombinant His-tagged CTF1 with native forms to rule out tag interference with protein function

  • Cell-type specific effects analysis: Systematically test CTF1 activity across different cell lineages, as receptor expression and downstream signaling machinery vary significantly between tissue types

  • Concentration-dependent response curves: Generate complete dose-response data as CTF1 may exhibit biphasic effects depending on concentration

  • Temporal analysis: Implement time-course experiments as CTF1 effects may vary significantly from acute to chronic exposure

  • Receptor complex characterization: Use co-immunoprecipitation with crosslinking to verify the composition of receptor complexes in your specific experimental system

  • Signaling pathway inhibition: Employ selective inhibitors to dissect which downstream pathways mediate observed effects

  • Validation across models: Compare in vitro findings with ex vivo and in vivo models when possible

This systematic approach helps identify experimental variables responsible for conflicting results and establishes which conditions reliably reproduce specific CTF1 functions.

What are the optimal experimental designs for studying CTF1's role in chromatin modification?

When investigating CTF1's chromatin-modifying capabilities, optimal experimental designs should incorporate:

  • Dual reporter systems: Implement systems containing telomere-proximal and telomere-distal reporter genes separated by CTF1 binding sites to simultaneously assess boundary effects, as demonstrated in published research

  • Mutational analysis: Compare wild-type CTF1 with mutants defective in either DNA binding or histone interaction to dissect domain-specific functions

  • Chromatin immunoprecipitation (ChIP): Perform ChIP analysis targeting various histone modifications (particularly acetylation) and variants (especially H2A.Z) to characterize the chromatin landscape

  • ChIP-seq approach: Expand beyond candidate loci to genome-wide analysis of CTF1-associated chromatin boundaries

  • Time-resolved ChIP: Assess the temporal establishment of boundaries following CTF1 expression or recruitment

  • Controlled protein recruitment: Use systems like GAL4-CTF1 fusion proteins that allow targeting to specific genomic loci via GAL4 binding sites

  • Correlation with transcriptional output: Integrate RNA-seq analysis to correlate boundary formation with gene expression changes

These approaches provide complementary data on CTF1's mechanism in establishing chromatin boundaries and preventing the spread of repressive chromatin marks from telomeres.

What are the optimal expression and purification methods for His-tagged CTF1?

For optimal expression and purification of His-tagged CTF1, the following methodology is recommended:

Expression System Selection:

  • Mammalian expression systems (HEK293 or CHO cells) are preferred for human CTF1 to ensure proper folding and post-translational modifications

  • For higher yield but potentially lower activity, bacterial systems (E. coli BL21(DE3)) with specifically optimized conditions can be considered

Expression Protocol:

  • Clone the human CTF1 coding sequence into a vector containing an N-terminal or C-terminal His-tag (N-terminal recommended to avoid interfering with receptor binding)

  • For mammalian expression, transfect cells and collect protein-containing media after 48-72 hours

  • For bacterial expression, induce with 0.5mM IPTG at lower temperatures (16-18°C) overnight to improve solubility

Purification Strategy:

  • Perform initial capture using Ni-NTA affinity chromatography (pH 8.0, 20mM imidazole in binding buffer, 250mM imidazole for elution)

  • Follow with size exclusion chromatography to remove aggregates and ensure monomeric protein

  • Confirm purity by SDS-PAGE (>95%) and verify identity by Western blot and/or mass spectrometry

  • Assess activity through in vitro cardiac myocyte hypertrophy assays or receptor binding studies

Storage Conditions:

  • Store purified protein at -80°C in small aliquots to avoid freeze-thaw cycles

  • Optimal buffer: 20mM Tris-HCl pH 7.5, 150mM NaCl, 10% glycerol

  • Stability testing should be performed to determine maximum storage time without activity loss

How can researchers effectively study the interaction between CTF1 and histones?

To effectively study CTF1-histone interactions, researchers should implement multiple complementary approaches:

In Vitro Binding Assays:

  • Employ purified His-tagged CTF1 and recombinant or purified histones (particularly H3.3 and H2A.Z) for direct binding assays

  • Use techniques such as microscale thermophoresis or isothermal titration calorimetry to determine binding affinities and thermodynamic parameters

  • Perform pull-down assays with immobilized His-tagged CTF1 to identify specific histone binding partners from nuclear extracts

Structural Characterization:

  • Identify the minimal histone-binding domain through truncation and mutation analysis

  • Consider X-ray crystallography or NMR spectroscopy of CTF1 complexed with histone peptides or nucleosomes

  • Use hydrogen exchange mass spectrometry to map interaction interfaces, similar to approaches used for other chromatin-binding factors

Cellular Assays:

  • Apply chromatin immunoprecipitation (ChIP) to assess CTF1 co-localization with specific histone modifications

  • Use proximity ligation assays to verify CTF1-histone interactions in situ

  • Implement FRET-based approaches with fluorescently tagged CTF1 and histones to study dynamic interactions

Functional Validation:

  • Construct CTF1 mutants with altered histone binding capacity and test their ability to establish chromatin boundaries

  • Assess histone modification patterns at CTF1-bound regions through sequential ChIP experiments

  • Correlate histone interaction strength with functional outcomes in gene expression studies

What control experiments are essential when studying CTF1's effect on gene expression?

When investigating CTF1's impact on gene expression, the following control experiments are essential:

Specificity Controls:

  • Include CTF1 binding site mutants that abolish DNA binding to confirm sequence-specific effects

  • Test CTF1 protein mutants lacking histone-binding capability to distinguish DNA-binding from chromatin-modifying functions

  • Employ unrelated DNA-binding proteins (e.g., GAL4-DBD alone) to rule out non-specific effects of protein recruitment

System Validation Controls:

  • Verify CTF1 expression levels through Western blotting and qPCR

  • Confirm CTF1 binding to target sites using ChIP-qPCR

  • Include positive controls known to affect gene expression (e.g., GAL4-VP16 fusion) to validate the experimental system

Experimental Design Controls:

  • Test effects at multiple genomic locations to distinguish position-specific from general effects

  • Include non-telomeric loci when studying boundary function to determine telomere specificity

  • Perform time-course experiments to distinguish direct from secondary effects

  • Assess concentration-dependent responses to determine optimal working concentrations

Technical Controls:

  • Use multiple independent reporter constructs to minimize clone-specific effects

  • Implement internal normalization strategies to control for transfection efficiency and cell number

  • When using inducible systems, verify background expression levels in uninduced conditions

  • Include vector-only controls to account for effects of transfection and selection

How should researchers analyze ChIP data to identify CTF1-mediated chromatin boundaries?

When analyzing ChIP data for CTF1-mediated chromatin boundaries, researchers should implement this comprehensive analytical framework:

Data Processing and Quality Control:

  • Assess sequencing quality metrics (Q scores, duplicate rates, alignment percentages)

  • Normalize for sequencing depth and input controls

  • Implement spike-in normalization when comparing conditions with potentially global changes

Boundary Identification:

  • Generate normalized signal tracks for histone modifications (particularly H3K9ac, H3K27ac, H4ac) and variants (H2A.Z)

  • Calculate the gradient of modification signals to identify transition zones

  • Correlate these transitions with CTF1 binding sites

  • Apply hidden Markov models or segmentation algorithms to systematically identify chromatin state transitions

Validation and Characterization:

  • Compare multiple histone modification patterns at identified boundaries

  • Determine the directionality and strength of the boundary effect

  • Calculate the distance over which the boundary effect extends

  • Compare telomeric versus non-telomeric CTF1 binding sites

Integrative Analysis:

  • Correlate boundary strength with gene expression changes (from RNA-seq)

  • Assess conservation of boundaries across cell types

  • Compare CTF1 boundaries with known insulators (CTCF, cohesins)

  • Integrate with three-dimensional chromatin interaction data (Hi-C, ChIA-PET)

Statistical Considerations:

  • Implement multiple testing correction for genome-wide analyses

  • Determine the significance of enrichment at boundaries versus matched control regions

  • Quantify boundary strength through modification gradient measurements

What statistical approaches are most appropriate for analyzing CTF1-induced cardiac hypertrophy?

When analyzing CTF1-induced cardiac hypertrophy data, researchers should utilize these statistical approaches:

Experimental Design Considerations:

  • Power analysis to determine sufficient biological replicates (minimum n=4 recommended)

  • Appropriate controls (vehicle, inactive CTF1 mutants, other cytokines)

  • Stratification of samples by relevant variables (cell source, culture conditions)

Primary Analysis for Cell Size Measurements:

  • Normalization to control conditions as fold-change or percentage increase

  • Assessment of normal distribution (Shapiro-Wilk test)

  • For normally distributed data: one-way ANOVA with post-hoc tests (Tukey or Dunnett)

  • For non-parametric data: Kruskal-Wallis with Dunn's post-hoc test

  • Consider mixed-effects models for repeated measures designs

Dose-Response Analysis:

  • Fit data to appropriate models (four-parameter logistic)

  • Calculate EC50 values and confidence intervals

  • Compare potency and efficacy parameters between experimental conditions

  • Test for biphasic responses that may indicate receptor complex heterogeneity

Time-Course Analysis:

  • Two-way ANOVA with time and treatment as factors

  • Area under the curve calculations for cumulative response

  • Regression analysis to determine response kinetics

  • Consider modeling using differential equations for complex pathways

Multivariate Approaches:

  • Principal component analysis for multiple hypertrophy markers

  • Hierarchical clustering to identify patterns across experimental conditions

  • Machine learning approaches for complex datasets integrating multiple endpoints

How can researchers effectively compare the chromatin boundary function of CTF1 with other boundary elements?

To effectively compare CTF1's chromatin boundary function with other boundary elements, researchers should implement this comparative framework:

Standardized Experimental Systems:

  • Design reporter systems where various boundary elements can be exchanged in the same genomic context

  • Implement dual reporter assays with a fixed distance between reporters

  • Generate isogenic cell lines with various boundaries integrated at identical genomic positions

Functional Metrics for Comparison:

  • Boundary strength: measured as the ratio of expression between protected and unprotected genes

  • Positional dependence: test function at varying distances from telomeres

  • Directionality: assess if protection extends bidirectionally or unidirectionally

  • Context dependence: test function across different cell types and chromatin environments

Mechanistic Characterization:

  • Compare histone modification profiles using standardized ChIP-seq protocols

  • Assess recruitment of chromatin remodeling complexes to different boundaries

  • Determine dependence on specific factors through depletion experiments

  • Analyze three-dimensional chromatin organization via 4C or Hi-C

Cross-boundary Comparison Table:

Boundary ElementDirectionalityHistone ModificationsRequired CofactorsTelomere SpecificityMechanism
CTF1UnidirectionalH3/H4 acetylation↑, H2A.Z recruitmentHistone acetyltransferasesYesHistone binding, H2A.Z recruitment
CTCFBidirectionalVariousCohesin complexNoLoop formation, topological domain boundary
tRNA genesBidirectionalH3K4meRNA Pol III machineryNoTranscriptional interference
[Other elements][Data][Data][Data][Data][Data]

Integration with Genomic Data:

  • Compare genome-wide binding profiles of different boundary elements

  • Analyze co-occurrence or mutual exclusion patterns

  • Correlate boundary strength with underlying sequence features

  • Assess evolutionary conservation of different boundary mechanisms

What are the promising approaches for investigating the dual functions of CTF1 in cardiac biology and chromatin regulation?

Investigating CTF1's dual roles in cardiac biology and chromatin regulation presents intriguing research opportunities through these integrative approaches:

Mechanistic Intersection Studies:

  • Determine if CTF1's chromatin regulatory function contributes to its cardiac effects by analyzing chromatin changes in cardiac myocytes treated with soluble CTF1

  • Develop CTF1 mutants with selective impairment of either cytokine signaling or chromatin binding to dissect pathway-specific effects

  • Investigate whether the same domains mediate both functions or if they operate independently

Genome-wide Approaches:

  • Perform integrated ChIP-seq and RNA-seq in cardiac cells to identify CTF1-regulated genes and associated chromatin changes

  • Employ CUT&RUN or CUT&Tag for higher resolution mapping of CTF1 chromatin interactions

  • Use ATAC-seq to assess global chromatin accessibility changes following CTF1 signaling activation

  • Apply single-cell multimodal approaches to correlate CTF1 activity with cell state transitions

Translational Research Directions:

  • Examine if chromatin-modifying abilities of CTF1 contribute to cardiac remodeling in disease models

  • Develop targeted CTF1 variants that selectively activate beneficial pathways while minimizing adverse effects

  • Investigate CTF1's potential epigenetic imprinting effects that may persist beyond acute signaling

Technological Innovations:

  • Apply CRISPR-based approaches to target CTF1 to specific genomic loci and assess localized chromatin effects

  • Develop optogenetic or chemically-inducible CTF1 systems for temporal control of activity

  • Employ organoid models to study CTF1 functions in three-dimensional tissue contexts

What technical challenges remain in studying CTF1-histone interactions and how might they be addressed?

Several technical challenges persist in studying CTF1-histone interactions, but emerging methodologies offer promising solutions:

Current Technical Limitations:

  • Transient nature of CTF1-histone interactions makes them difficult to capture

  • Potential for artifacts in overexpression systems

  • Difficulty distinguishing direct from indirect interactions

  • Limited resolution of traditional ChIP approaches

  • Challenges in reconstituting physiologically relevant chromatin in vitro

Innovative Solutions:

  • Proximity labeling approaches: Implement BioID or APEX2 fusions with CTF1 to biotinylate nearby proteins, enabling identification of transient histone interactions

  • Live-cell single-molecule tracking: Apply techniques like SPT-PALM to visualize CTF1-chromatin interactions in real-time

  • In situ protein interaction visualization: Employ proximity ligation assays or fluorescence complementation to detect interactions in cellular contexts

  • Advanced ChIP techniques: Utilize CUT&RUN, CUT&Tag, or ChEC-seq for higher resolution and lower background

  • Cryo-EM approaches: Apply to larger CTF1-nucleosome complexes to obtain structural insights

Cross-disciplinary Strategies:

  • Biophysical approaches: Apply hydrogen-deuterium exchange mass spectrometry similar to methods used for Hsf1

  • Computational modeling: Implement molecular dynamics simulations to predict interaction interfaces

  • Synthetic biology: Design minimal systems to test specific aspects of CTF1-histone interactions

  • Chemical biology: Develop photo-crosslinking approaches to capture transient interactions

Data Integration Frameworks:

  • Triangulate results from multiple complementary techniques

  • Develop standardized pipelines for comparing results across experimental systems

  • Establish open databases for CTF1-chromatin interaction data sharing

How might understanding CTF1's dual functions impact therapeutic approaches to cardiac diseases?

Understanding CTF1's dual functions as both a secreted cytokine and a chromatin regulatory factor opens novel therapeutic avenues for cardiac diseases through multiple mechanistic pathways:

Targeted Therapeutic Strategies:

  • Development of CTF1 variants with enhanced chromatin boundary function but reduced pro-hypertrophic signaling for selective treatment of specific cardiac conditions

  • Design of small molecules that modulate CTF1's interaction with either receptors or chromatin to achieve pathway-specific effects

  • Exploration of CTF1-based gene therapy approaches utilizing its boundary function to ensure stable long-term transgene expression in cardiac tissue

Disease-Specific Applications:

  • For pathological hypertrophy: Selective inhibition of CTF1's cytokine signaling while preserving beneficial chromatin functions

  • For heart failure: Targeted delivery of modified CTF1 to improve cardiomyocyte survival through specific epigenetic modifications

  • For cardiac regeneration: Exploitation of CTF1's ability to create favorable chromatin environments for cardiac progenitor gene expression

Diagnostic and Prognostic Implications:

  • Development of biomarkers based on CTF1-regulated epigenetic signatures

  • Stratification of cardiac patients based on CTF1 pathway activity profiles

  • Personalized medicine approaches targeting specific aspects of CTF1 signaling based on individual patient epigenetic landscapes

Product Science Overview

Molecular Characteristics
  • Amino Acid Sequence: Cardiotrophin-1 consists of 201 amino acids. The recombinant version, often produced in human 293 cells (HEK293), includes a polyhistidine tag (His Tag) at the N-terminus .
  • Molecular Weight: The protein has a calculated molecular weight of approximately 23 kDa. However, due to glycosylation, it migrates as 27-30 kDa on SDS-PAGE .
  • Purity: The recombinant protein is typically purified to greater than 90% purity as determined by SDS-PAGE and SEC-MALS .
Biological Functions

Cardiotrophin-1 is known for its ability to:

  • Induce Cardiac Hypertrophy: It stimulates the hypertrophic growth of cardiac myocytes, which is a critical response during heart development and in certain pathological conditions .
  • Activate Signaling Pathways: CT-1 binds to and activates the ILST/gp130 receptor, which in turn activates the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. This signaling cascade is essential for transducing hypertrophic and cytoprotective signals in cardiac cells .
  • Neurotrophic Functions: CT-1 also has neurotrophic properties, supporting the survival of motoneurons in the spinal cord and brainstem .
  • Hepatocyte Survival: It acts as a hepatocyte survival factor, reducing liver cell damage in models of acute liver injury .
Clinical Relevance
  • Heart Failure: Elevated levels of Cardiotrophin-1 have been observed in patients with heart failure, dilated cardiomyopathy, and other cardiac conditions .
  • Protective Role: CT-1 expression increases in response to hypoxic conditions, providing protection to cardiac cells during ischemic events .
Recombinant Production
  • Expression System: The recombinant form of Cardiotrophin-1 is often produced in Escherichia coli or human 293 cells (HEK293) .
  • Formulation: The protein is typically lyophilized from a filtered solution and can be reconstituted in sterile water or other suitable buffers .
  • Storage: For long-term storage, the lyophilized protein should be kept at -20°C or lower. Once reconstituted, it should be stored at 4°C for short-term use and at -70°C for long-term storage .

Cardiotrophin-1 (Human Recombinant, His Tag) is a valuable tool in research due to its significant roles in cardiac hypertrophy, neuroprotection, and hepatocyte survival. Its recombinant form allows for detailed studies and potential therapeutic applications.

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 2024 Thebiotek. All Rights Reserved.