The term "CTF2P Mouse" refers to a potential misinterpretation of nomenclature. CTF2P is a human pseudogene (cardiotrophin 2, pseudogene), while its functional counterpart in mice is CTF2 (cardiotrophin-2), encoding the cytokine neuropoietin (NP). This distinction is critical for understanding its biological roles and evolutionary divergence between species.
Species | Gene Name | Status | Protein |
---|---|---|---|
Human | CTF2P | Pseudogene | Non-functional |
Mouse | CTF2 | Functional | Neuropoietin (NP) |
Amino Acid Sequence: The mature protein (Ala23-Ala204) includes a 22 aa signal peptide and a 182 aa mature segment .
Receptor Binding: Neuropoietin signals via a receptor complex comprising gp130, CNTF receptor-α (CNTFRα), and leukemia inhibitory factor receptor (LIFR) .
Human Pseudogene Formation: An 8 bp deletion in the human CTF2P disrupts the reading frame, rendering it non-functional. This loss is compensated by CNTF signaling .
Mouse Functional Conserved: CTF2 remains intact, enabling neuropoietin’s role in motor neuron and neural precursor survival .
Recombinant mouse neuropoietin (e.g., R&D Systems’ 2709-NP, Thermofisher’s 250-25B-25UG) is used in:
Application | Parameters | Source |
---|---|---|
Bioassays | ED₅₀: 50–200 ng/mL for neuronal survival | |
Protein Stability | Lyophilized with BSA (carrier) or PBS (carrier-free) |
Feature | CTF2 (Mouse) | CNTF | CT-1 |
---|---|---|---|
Receptor | gp130 + CNTFRα + LIFR | gp130 + CNTFRα + LIFR | gp130 + LIFR + WSX-1 |
Primary Role | Embryonic motor neuron survival | Postnatal motor neuron maintenance | Cardiac and skeletal muscle development |
Expression | Embryonic neuroepithelia | Neurons, glia | Heart, liver, skeletal muscle |
CTF2P, also known as Neuropoietin, belongs to the IL-6 family of cytokines. It is the result of a gene duplication involving cardiotrophin-1 (CT-1) and is part of a subfamily within the IL-6 family that includes CT-1, CLC, and CTNF. CTF2P contributes to an increase in platelet count, often associated with splenomegaly, and plays a role in the development and maturation of neuronal precursors.
Recombinant Mouse Neuropoietin, produced in E. coli, is a polypeptide chain that is not glycosylated. It consists of 182 amino acids, resulting in a molecular weight of approximately 19.7 kDa. The purification of CTF2P is achieved through specialized chromatographic methods.
Sterile, white powder that has been lyophilized (freeze-dried) and filtered.
The protein is lyophilized from a concentrated solution (0.2 μm filtered) containing 20mM Tris-HCl at pH 8.0, 0.5mM DTT, and 500mM NaCl.
For reconstitution of the lyophilized Neuropoietin, it is recommended to use sterile PBS at a concentration of at least 100 μg/ml. This solution can be further diluted with other aqueous solutions as needed.
Lyophilized CTF2P remains stable at room temperature for up to 3 weeks; however, it is recommended to store it desiccated at a temperature below -18°C. Once reconstituted, Neuropoietin should be stored at 4°C for a period of 2-7 days. For long-term storage, it is advisable to store it at -18°C. Avoid repeated freeze-thaw cycles.
The purity of the product is determined using the following methods and is guaranteed to be greater than 98.0%:
(a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis.
(b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
The ED50, determined by a cell proliferation assay using human TF-1 cells, is less than 200 ng/ml. This corresponds to a specific activity greater than 5000 IU/mg.
Cardiotrophin-2, CT-2, Neuropoietin, Np, Ctf2, Gm494.
Escherichia Coli.
APISPSEPIG QAYSLALYMQ KNTSALLQTY LQHQGSPFSD PGFSAPELQL STLPSAAVSF KTWHAMEDAE RLSRAQGAFL ALTQHLQLVG DDQSYLNPGS PILLAQLGAA RLRAQGLLGN MAAIMTALGL PIPPEEDTLG FVPFGASAFE RKCRGYIVTR EYGHWTDRAV RDLALLKAKY SA.
Neuropoietin (NP), encoded by the CTF2 gene in mice, is a 22 kDa member of the IL-6 family of cytokines. In mice, it is synthesized as a 204 amino acid precursor that contains a 22 amino acid signal sequence and a 182 amino acid mature segment. The secreted molecule is characterized by the presence of four alpha-helices, typical of hematopoietic superfamily molecules .
Mouse Neuropoietin signals through a receptor complex comprising the CNTF receptor alpha (CNTFRα) component, glycoprotein 130 (gp130), and Leukemia Inhibitory Factor Receptor (LIFR) . This is the same receptor complex utilized by CNTF, which explains why CNTF signaling can likely compensate for the absence of functional Neuropoietin in humans. The signaling pathway typically activates the JAK/STAT pathway, particularly STAT3, which regulates gene expression related to neuronal development and survival .
Neuropoietin is predominantly expressed in mouse neuroepithelia during embryonic development . It appears to be selectively expressed in tissues involved with nervous system development during embryogenesis . Unlike some other cytokines that show broad expression patterns across multiple tissues, Neuropoietin exhibits a more restricted expression profile, highlighting its specialized role in neural development . This developmental-stage specific expression pattern suggests careful temporal regulation of the gene, which is critical for proper nervous system development.
For accurate detection and quantification of Neuropoietin in mouse tissues, researchers should employ a multi-platform approach:
RT-qPCR analysis: Design primers specific to unique regions of the CTF2 transcript that do not cross-react with related cytokines (especially CT-1). Include appropriate housekeeping genes for normalization, and validate primer specificity using gel electrophoresis of PCR products.
Immunohistochemistry/Immunofluorescence: Utilize validated antibodies such as biotinylated antibodies against mouse Neuropoietin (e.g., those recognizing the Ala23-Ala204 region) . When performing these analyses, include appropriate negative controls (secondary antibody only) and positive controls (tissues known to express Neuropoietin).
Western blotting: For protein detection, use denaturing conditions that can distinguish Neuropoietin from other IL-6 family members based on molecular weight (approximately 22 kDa) .
RNA-Seq: For comprehensive transcriptomic analysis, ensure sufficient sequencing depth (>30 million reads per sample) to detect potentially low-abundance transcripts like Neuropoietin.
Optimal storage conditions for antibodies against mouse Neuropoietin include storage at -20 to -70°C for up to 12 months from receipt date, 2-8°C under sterile conditions after reconstitution for up to 1 month, or -20 to -70°C under sterile conditions after reconstitution for up to 6 months .
To investigate Neuropoietin's role in motor neuron survival, researchers should consider these methodological approaches:
Primary motor neuron cultures: Isolate embryonic spinal motor neurons and treat with recombinant Neuropoietin at varying concentrations (1-100 ng/mL). Assess survival using TUNEL assay, caspase-3 activation, or live/dead staining over 3-7 days in culture.
CTF2 knockout models: Generate conditional knockout mice using Cre-loxP systems to delete CTF2 in specific tissues or developmental stages. This approach is essential since Neuropoietin's embryonic expression suggests potential developmental roles that might be missed in constitutive knockouts.
Receptor blocking studies: Use neutralizing antibodies or soluble receptor fragments to block specific components of the Neuropoietin receptor complex (CNTFRα, gp130, LIFR) to determine which signaling components are essential for motor neuron survival effects .
Molecular signaling analysis: Examine phosphorylation of STAT3, ERK1/2, and AKT following Neuropoietin treatment to delineate the specific intracellular pathways mediating survival effects.
In vivo neuronal survival assays: Following axotomy or neurotoxin challenge, administer recombinant Neuropoietin and quantify motor neuron survival compared to vehicle control.
These approaches should be complemented with appropriate controls, including other IL-6 family cytokines like CNTF and CT-1 for comparison of potency and specificity.
This represents a complex research challenge requiring several specialized approaches:
Comparative genomics analysis: Perform detailed sequence alignment of the CTF2/CTF2P locus across multiple mammalian species to identify when the 8 base pair deletion occurred in the evolutionary timeline. Tools like MEGA, PAML, or PhyloP can help determine whether the pseudogenization event was under positive selection .
Transgenic humanized mouse models: Create mouse models where the functional mouse CTF2 is replaced with the human CTF2P pseudogene to observe phenotypic consequences. This allows assessment of whether CNTF or other cytokines genuinely compensate for Neuropoietin absence in a mammalian system.
Functional complementation assays: In cell culture systems lacking both CNTF and Neuropoietin, introduce either mouse Neuropoietin or human CNTF to determine if they rescue identical downstream signaling events and biological outcomes.
Receptor occupancy and competition studies: Using surface plasmon resonance or similar techniques, examine whether human CNTF and mouse Neuropoietin compete for the same binding sites on receptor complexes with similar affinities, supporting the compensation hypothesis .
Single-cell transcriptomics: Compare expression patterns in homologous neural tissues between mice and humans to identify potential compensatory mechanisms in human cells that might explain why CTF2P pseudogenization was tolerated during evolution.
This multi-faceted approach allows researchers to understand the evolutionary and functional implications of CTF2P pseudogenization in humans while leveraging the mouse model for mechanistic insights.
The compensation hypothesis is supported by several lines of evidence:
Receptor complex sharing: Both Neuropoietin and CNTF signal through an identical receptor complex comprising CNTFRα, gp130, and LIFR, suggesting functional redundancy .
Evolutionary conservation analysis: The pseudogenization of CTF2P occurred in the human lineage without apparent negative selection, suggesting its function became dispensable. This timing correlates with evolutionary changes in the CNTF locus that may have enhanced its compensatory capacity .
Functional similarity: Both Neuropoietin and CNTF promote motor neuron survival and can increase the proliferation of neural precursor cells, indicating overlapping biological functions .
Expression pattern complementarity: In mice, CNTF expression increases in regions where Neuropoietin expression diminishes during later developmental stages, suggesting a temporal handoff of function .
Interspecies conservation patterns: While human CTF2P is a pseudogene, the sequence homology of mouse Neuropoietin remains high across other species (95% amino acid identity with rat, 90% with dog, and 88% with chimpanzee), indicating functional importance in these species .
The evidence collectively suggests that CNTF likely assumed the functions of Neuropoietin in humans through a process of evolutionary compensation, allowing the CTF2 gene to degenerate into a pseudogene without detrimental effects.
Neuropoietin shares several structural features with other IL-6 family members while maintaining distinct characteristics:
Four-helix bundle structure: Like other IL-6 family cytokines, Neuropoietin contains four alpha-helices arranged in the typical up-up-down-down topology characteristic of this family .
Evolutionary relationship: Neuropoietin appears to be the product of a gene duplication event involving cardiotrophin-1 (CT-1), defining a subfamily within the IL-6 family that includes CT-1, CLC (cardiotrophin-like cytokine), and CNTF .
Signal sequence: Mouse Neuropoietin contains a 22 amino acid signal sequence that facilitates secretion, similar to other secreted cytokines in this family .
Receptor binding interfaces: Neuropoietin contains specific binding sites for CNTFRα that are analogous to those in CNTF, allowing both cytokines to compete for the same receptor .
Species-specific conservation: Unlike some highly conserved cytokines, Neuropoietin shows variable conservation across species, with the human version having evolved into a pseudogene while maintaining high sequence homology among rodent species (95% amino acid identity between mouse and rat) .
These structural features explain both the functional overlap with other IL-6 family cytokines (particularly CNTF) and Neuropoietin's unique developmental roles in neural tissues.
Detecting mouse Neuropoietin using antibody-based methods presents several technical challenges:
Cross-reactivity concerns: Due to high sequence similarity with other IL-6 family cytokines (particularly CT-1), antibodies may cross-react with related proteins. Solution: Validate antibody specificity using knockout tissues or overexpression systems, and perform pre-absorption controls with recombinant related cytokines.
Low endogenous expression levels: Neuropoietin is often expressed at low levels, making detection difficult. Solution: Consider signal amplification methods such as tyramide signal amplification for immunohistochemistry or chemiluminescent substrate optimization for Western blots .
Temporal expression limitations: Since Neuropoietin is predominantly expressed during embryonic development, timing of tissue collection is critical. Solution: Carefully stage embryos based on established criteria (somite number, etc.) and process tissues immediately to prevent protein degradation.
Antibody storage and handling: Improper storage can lead to antibody degradation and false-negative results. Solution: Store according to manufacturer recommendations: -20 to -70°C for long-term storage, and avoid repeated freeze-thaw cycles .
Protein conformation dependency: Some antibodies recognize only specific conformations of Neuropoietin. Solution: Use multiple antibodies targeting different epitopes, and optimize fixation conditions (paraformaldehyde vs. methanol) for immunohistochemistry applications.
For optimal results with biotinylated antibodies against mouse Neuropoietin, researchers should determine optimal dilutions for each application through titration experiments and include appropriate positive and negative controls in every experiment .
Distinguishing Neuropoietin-specific effects from those of related cytokines requires careful experimental design:
Receptor component blocking: Selectively block specific receptor components using neutralizing antibodies or soluble receptor fragments. While both CNTF and Neuropoietin use CNTFRα, gp130, and LIFR, subtle differences in binding kinetics can be exploited .
Dose-response relationships: Systematically compare dose-response curves of Neuropoietin versus CNTF and CT-1. Differences in EC50 values can help distinguish their relative potencies in specific assays.
Temporal activation patterns: Monitor the kinetics of downstream signaling activation (STAT3, ERK1/2, AKT phosphorylation) following stimulation. Neuropoietin may induce different temporal patterns of activation compared to other family members.
Genetic approaches: Use CRISPR/Cas9 to create cell lines specifically lacking CTF2 while maintaining expression of other IL-6 family cytokine genes, allowing clean assessment of Neuropoietin-specific functions.
Receptor complex assembly analysis: Employ proximity ligation assays or FRET-based approaches to visualize receptor complex formation kinetics, which may differ between Neuropoietin and other cytokines despite using the same components .
Transcriptional profiling: Perform RNA-Seq following treatment with different IL-6 family cytokines to identify Neuropoietin-specific gene expression signatures that distinguish its effects from those of CNTF or CT-1.
These approaches, used in combination, can help researchers attribute observed biological effects specifically to Neuropoietin rather than to related cytokines signaling through overlapping receptor complexes.
Several innovative research directions emerge from this evolutionary divergence:
Compensatory mechanism characterization: Investigate precisely how CNTF and potentially other cytokines compensate for the lack of functional Neuropoietin in humans. This could reveal principles of signaling network robustness applicable to other biological systems .
Therapeutic implications: Explore whether recombinant Neuropoietin could offer therapeutic advantages over CNTF in treating motor neuron diseases. Mouse Neuropoietin might have unique properties that were lost in human evolution but could be reintroduced therapeutically.
Evolutionary medicine: Study the CTF2P pseudogenization event in the context of other genetic changes in human evolution to understand how complex signaling networks adapt when key components are lost .
Neural development comparisons: Examine how the absence of functional Neuropoietin might contribute to subtle differences in neural development between humans and other mammals, particularly in motor neuron connectivity patterns.
Restoration experiments: Using gene editing techniques, restore functional CTF2 expression in human neural cells and examine the consequences for development, survival, and function. This could reveal whether the pseudogenization event had previously unrecognized beneficial effects.
These research directions would not only enhance our understanding of Neuropoietin biology but also provide insights into principles of molecular evolution, redundancy in signaling networks, and potential therapeutic approaches for neurological disorders.
Mouse neuropoietin is synthesized as a 204 amino acid precursor that contains a 22 amino acid signal sequence and a 192 amino acid mature segment . The secreted molecule is characterized by the presence of four alpha-helices, typical of hematopoietic superfamily molecules . The recombinant form of mouse neuropoietin is typically produced in E. coli and is available in both carrier-free and carrier protein formulations .
Recombinant mouse neuropoietin is used in various research applications, including cell culture and ELISA standards. It is typically lyophilized from a filtered solution and can be reconstituted in sterile PBS or aqueous buffer containing 0.1% BSA . For optimal stability, it should be stored at -20 to -70°C and should avoid repeated freeze-thaw cycles .