Cardiotrophin 1 (CTF1) is a secreted cytokine encoded by the CTF1 gene (NCBI Gene ID: 1489), primarily associated with cardiac hypertrophy and immune modulation.
In mammalian cells, CTF1 (distinct from cardiotrophin 1) is a transcription factor belonging to the CTF/NFI family, critical for chromatin domain boundary formation and gene silencing prevention.
Entity | Gene Symbol | Primary Function | Key References |
---|---|---|---|
Cardiotrophin 1 | CTF1 | Cytokine, cardiac hypertrophy | |
Transcription Factor | (Unresolved) | Chromatin boundary, epigenetic control |
Note: The transcription factor CTF1 described in telomere studies may belong to the CTF/NFI family (e.g., NFIA, NFIB), but its specific gene symbol remains unconfirmed in human contexts. Researchers should verify gene identifiers in publications.
Aspect | Cardiotrophin 1 | Transcription Factor CTF1 |
---|---|---|
Molecular Class | Cytokine | DNA-binding protein |
Cellular Targets | Cardiomyocytes, leukemic cells | Telomeric chromatin, histones |
Disease Links | Cardiomyopathy, leukemia | Gene silencing, telomere dysfunction |
Therapeutic Potential | Cardiac remodeling, oncology | Epigenetic therapy, transgene stability |
CTF1 Nomenclature: Resolve ambiguity in gene symbols for the transcription factor.
Cardiotrophin 1: Investigate its dual roles in cardiac hypertrophy and leukemia.
Epigenetic Regulation: Explore whether CTF1-mediated H2A.Z recruitment is conserved across species.
CTF1, also known as CT-1 or CT1, is a member of the interleukin-6 family of cytokines with significant expression in tissues critically involved in metabolic regulation, including beta-cells, skeletal muscle, and liver . Located on chromosome 16p11.2 (at position 30,896,607 bp to 30,903,560 bp on the plus strand), the CTF1 gene encodes cardiotrophin 1, which functions in multiple biological processes .
From a molecular perspective, CTF1 engages in numerous cellular activities including:
Cell surface receptor signaling pathways
Cell-cell signaling
Nervous system development
Muscle organ development
Cell proliferation regulation
Positive regulation of tyrosine phosphorylation of Stat3 protein
CTF1 contains specific domains that determine its diverse functions. The protein includes a DNA-binding domain and a histone-binding domain, particularly its proline-rich domain which has been shown to interact with histone H3.3 . This dual functionality allows CTF1 to act both as a transcriptional regulator and as a modulator of chromatin structure.
The proline-rich domain specifically mediates transcriptional activation in response to growth factors in mammalian cells through its interaction with histone H3.3 . This structural feature is crucial for researchers studying the mechanistic basis of CTF1's effects on gene expression and chromatin organization.
When investigating CTF1 function, researchers should consider several experimental approaches depending on the specific aspects being studied:
For chromatin regulation studies:
Chromatin immunoprecipitation (ChIP) assays have successfully demonstrated CTF1's ability to establish boundaries between differentially acetylated chromatin domains
Reporter gene assays using dual reporter systems where telomere-distal and telomere-proximal loci are monitored simultaneously
For metabolic function studies:
Hyperinsulinemic-euglycemic clamp studies provide the gold standard for assessing insulin sensitivity in vivo
Oral glucose tolerance tests with derived indices of insulin sensitivity and secretion
Magnetic resonance imaging/spectroscopy for tissue-specific effects
For genetic studies:
SNP genotyping focusing on tagging SNPs such as rs1046276, rs1458201, and rs8046707, which capture common genetic variation in the CTF1 locus
CTF1 has been linked to aging processes primarily through its effects on vascular stiffness and fibrosis. Mice lacking CTF1 display decreased arterial stiffness, develop less vascular fibrosis, and live longer than wild-type animals . These observations suggest CTF1 may promote age-related vascular dysfunction, a key component of human aging.
The inclusion of CTF1 in the GenAge database (HAGRID: 304) indicates its relevance to aging research, although the evidence is described as "indirect or inconclusive" for human aging specifically . This classification suggests that while animal model data is compelling, direct human evidence requires further investigation.
Methodologically, researchers investigating CTF1's role in human aging should consider:
Longitudinal studies correlating CTF1 expression with age-related vascular parameters
Examination of CTF1 genetic variants in population cohorts with well-characterized longevity
In vitro studies using senescent human cells to assess CTF1's effects on senescence-associated secretory phenotype
To investigate CTF1's impact on age-related phenotypes, researchers should employ multiple complementary approaches:
For vascular aging assessment:
Pulse wave velocity measurements as a clinical gold standard for arterial stiffness
Histological analysis of arterial samples for fibrosis quantification
Expression profiling of extracellular matrix components in vascular tissues
For cellular senescence studies:
Senescence-associated β-galactosidase assays in CTF1-treated or CTF1-deficient cells
Analysis of senescence markers (p16, p21) in response to CTF1 manipulation
Assessment of telomere length dynamics in the presence of varying CTF1 levels
For epigenetic aging analysis:
DNA methylation age calculation using established epigenetic clocks
Histone modification patterns at age-regulated genes in response to CTF1
Researchers should be aware that CTF1's effects may be tissue-specific, requiring careful selection of experimental systems relevant to the aging phenotype under investigation.
A study of 1,771 German subjects demonstrated that genetic variation in the CTF1 locus significantly impacts insulin sensitivity. Specifically, the minor allele of SNP rs8046707 was associated with decreased in vivo measures of insulin sensitivity after appropriate statistical adjustment . This finding contrasts with mouse studies suggesting that CTF1 improves insulin sensitivity, highlighting important species differences.
Table 1: Key CTF1 SNPs and their metabolic associations
SNP ID | Minor Allele Frequency | Observed Effect | Association Strength |
---|---|---|---|
rs8046707 | 0.41 | Decreased insulin sensitivity | Significant |
rs1458201 | 0.25 | Increased VLDL levels | Nominal |
rs1046276 | 0.36 | No significant metabolic effects | N/A |
The genetic linkage between these SNPs was modest: r² = 0.60 between rs1046276 and rs1458201, r² = 0.38 between rs1046276 and rs8046707, and r² = 0.23 between rs1458201 and rs8046707 . This relatively weak linkage suggests these variants may have independent functional effects.
When investigating CTF1's role in glucose metabolism, researchers should consider:
For in vivo human studies:
Hyperinsulinemic-euglycemic clamp studies remain the reference standard for measuring insulin sensitivity
Oral glucose tolerance tests with calculation of insulin sensitivity indices (e.g., Matsuda index)
Continuous glucose monitoring for dynamic assessment of glycemic variability
Tracer studies to measure hepatic glucose production and peripheral glucose disposal
For molecular mechanism studies:
For pancreatic β-cell studies:
Glucose-stimulated insulin secretion assays in isolated islets with CTF1 treatment
Assessment of β-cell apoptosis markers following CTF1 exposure
Calcium imaging to evaluate β-cell function in response to CTF1
The relationship between CTF1 and adipose tissue distribution provides important insights into its metabolic effects. In magnetic resonance imaging studies, the minor A-allele of SNP rs8046707 was nominally associated with reduced visceral adipose tissue (VAT) . This finding is particularly significant given that:
Visceral adiposity is more strongly associated with insulin resistance than subcutaneous fat
The same allele was associated with decreased insulin sensitivity, creating an apparent paradox
This suggests CTF1 may affect insulin sensitivity through mechanisms independent of visceral fat accumulation
Researchers investigating these relationships should consider:
Adipose tissue depot-specific expression analysis of CTF1 and its receptors
Lipolysis and lipogenesis assays in different fat depots with CTF1 treatment
Assessment of adipose tissue inflammation markers in relation to CTF1 signaling
CTF1 exhibits significant activity as a chromatin domain boundary protein, protecting genes from telomeric silencing effects. Research has demonstrated that CTF1 can prevent the silencing of telomere-distal transgenes when its DNA-binding sites are positioned between the gene and the telomeric extremity .
The mechanism of this boundary function involves:
Recruitment of the histone variant H2A.Z to the telomeric locus
Restoration of high histone acetylation levels to the insulated telomeric transgene
Demarcation of chromatin structures with distinct histone acetylation status
For researchers studying CTF1's boundary function, chromatin immunoprecipitation (ChIP) experiments have proven particularly valuable, showing how CTF1 can establish boundaries between differentially acetylated chromatin domains.
To study CTF1's interactions with chromatin effectively, researchers should consider:
For examining histone interactions:
Co-immunoprecipitation assays to confirm CTF1's interaction with histone H3.3
In vitro binding assays with recombinant CTF1 domains and histone proteins
Mutational analysis of CTF1's proline-rich domain to identify critical residues for histone binding
For assessing boundary function:
Dual reporter gene assays where expression of telomere-proximal and telomere-distal loci can be monitored simultaneously
CRISPR-mediated deletion or insertion of CTF1 binding sites at endogenous boundary regions
Chromatin conformation capture techniques (3C, 4C, Hi-C) to assess long-range chromatin interactions
For analyzing histone modifications:
ChIP-seq for histone modifications (H3K9ac, H3K27ac, H3K9me3) in the presence or absence of CTF1
CUT&RUN or CUT&Tag for high-resolution mapping of CTF1 binding and associated histone modifications
Sequential ChIP to determine co-occurrence of CTF1 and specific histone modifications
These approaches should be complemented by functional assays to determine the biological consequences of CTF1's chromatin interactions.
CTF1's capacity to establish chromatin boundaries offers valuable applications in experimental systems:
For transgene expression stabilization:
For engineered gene regulation:
CTF1 binding sites can be used to insulate promoters from enhancers or silencers
Fusion proteins containing CTF1's histone-binding domain can be targeted to specific genomic loci to modify local chromatin structure
For studying telomeric silencing:
CTF1-based reporter systems can serve as sensors for telomeric heterochromatin spreading
This allows investigation of factors affecting telomere position effect in human cells
When designing such experimental systems, researchers should be aware that the histone-binding function of CTF1 is critical—protein fusions containing CTF1's histone-binding domain displayed similar boundary activities, while mutants impaired in histone interaction did not .
Developing CTF1-targeted therapeutics presents several challenges that researchers must address:
Contradictory functional effects:
While CTF1 knockout in mice leads to increased longevity and decreased vascular stiffness , the same mechanism could potentially compromise cardiac repair after injury
Human genetic data suggests CTF1 variants associated with decreased insulin sensitivity , contradicting mouse studies showing CTF1 improves insulin sensitivity
Tissue-specific effects:
CTF1 functions differently across tissues, requiring targeted delivery systems for therapeutic applications
The complex interplay between CTF1's metabolic and epigenetic functions complicates intervention design
Methodological considerations for therapeutic development:
Development of highly specific CTF1 agonists/antagonists that don't affect other IL-6 family cytokines
Establishment of appropriate biomarkers to monitor CTF1 activity in clinical settings
Design of delivery systems capable of targeting specific tissues where CTF1 modulation would be beneficial
Researchers pursuing CTF1-based therapeutics should consider combinatorial approaches that can address these complexities, potentially involving tissue-specific delivery of CTF1 modulators alongside complementary agents that address downstream pathway components.
The complexity of CTF1 function calls for integrative approaches combining multiple data types:
Recommended multi-omics strategy:
Integration of genomics (CTF1 genetic variants), transcriptomics (expression patterns), and epigenomics (histone modifications, chromatin structure)
Correlation with proteomics to assess post-translational modifications of CTF1 and interacting partners
Metabolomics to capture downstream metabolic effects of CTF1 signaling
Data integration methods:
Network analysis approaches linking CTF1 to broader signaling networks
Machine learning algorithms to identify patterns in multi-dimensional data
Pathway enrichment analysis to contextualize CTF1 effects within biological systems
Single-cell approaches:
Single-cell RNA-seq to identify cell populations particularly responsive to CTF1
Single-cell ATAC-seq to examine chromatin accessibility changes in response to CTF1
Spatial transcriptomics to map CTF1 activity in tissue contexts
These integrative approaches can help reconcile apparently contradictory findings and provide a more comprehensive understanding of CTF1's biological roles.
Several cutting-edge technologies offer significant potential for advancing CTF1 research:
Genome editing technologies:
CRISPR-based epigenome editing to modulate CTF1 expression in specific tissues
Base editing or prime editing to introduce or correct specific CTF1 variants
CRISPR activation/inhibition systems for temporally controlled CTF1 modulation
Advanced imaging approaches:
Live-cell imaging of CTF1-chromatin interactions using fluorescently tagged proteins
Super-resolution microscopy to visualize chromatin boundary formation
Intravital microscopy to observe CTF1 activity in living tissues
Organoid and tissue engineering systems:
Multi-lineage organoids to study CTF1's effects on tissue development and function
Engineered tissues with controllable CTF1 expression for aging and metabolic studies
Microfluidic organ-on-chip systems to examine CTF1's effects on tissue-tissue interactions
These technologies will allow researchers to address complex questions about CTF1 function with unprecedented precision and physiological relevance.
CT-1 is a 201 amino acid protein with a molecular mass of approximately 21 kDa . The recombinant form of CT-1 is typically produced in Escherichia coli (E. coli) and is available in both glycosylated and non-glycosylated forms . The protein is often lyophilized and can be reconstituted in sterile solutions for various applications .
CT-1 is a pleiotropic cytokine, meaning it has multiple effects on different cell types . It activates gp130-dependent signaling and stimulates the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway . This signaling pathway is crucial for transducing hypertrophic and cytoprotective signals in cardiac myocytes . Additionally, CT-1 has neurotrophic functions and is involved in the survival of motoneurons .
CT-1 expression is augmented after hypoxic stimulation and can protect cardiac cells during ischemic events . It has been shown to induce the expression of protective heat shock proteins (hsps) in cardiac cells . Elevated levels of CT-1 have been observed in patients with heart failure, dilatative cardiomyopathy, mitral regurgitation, stable and unstable angina, and after acute myocardial infarction .
Recombinant human CT-1 is used in various research applications, including: