Activates B cells via gp130 receptor signaling, promoting hyperplasia and autoimmunity .
Enhances myeloid differentiation and hematopoietic progenitor cell expansion in bone marrow .
Critical for motor neuron survival; knockout mice exhibit neonatal lethality due to suckling defects .
Suppresses thermogenesis in brown adipose tissue by inhibiting PGC-1α/β via CNTFR-STAT3 signaling .
Elevated in obesity, contributing to metabolic dysfunction .
Crisponi/Cold-Induced Sweating Syndrome (CISS2): Autosomal recessive mutations cause hyperthermia, skeletal anomalies, and cold-induced sweating .
Disease | Mechanism | Clinical Impact |
---|---|---|
CISS2 | Impaired CRLF1/CLCF1/CNTFRα signaling | Neonatal hyperthermia, dysautonomia |
Glioma | STAT3-mediated immunosuppression | Reduced survival, therapeutic target |
Obesity | Suppressed brown fat thermogenesis | Insulin resistance, metabolic syndrome |
Thermogenesis Regulation (2023):
Glioma Prognosis (2022):
Hematopoiesis (ProSpec Bio, 2022):
CLCF1 is an IL-6 family cytokine with neurotrophic and immuno-modulating functions. It plays significant roles in various biological processes including adipocyte thermogenesis regulation, hematopoiesis with a myeloid bias, and potential involvement in cancer progression. In brown adipose tissue, CLCF1 signaling acts as a brake on thermogenesis, which becomes dysregulated in obesity conditions. The cytokine was initially identified for its neurotrophic properties, but subsequent research has revealed its multifaceted roles in immune regulation and metabolic homeostasis .
Flow cytometry using monoclonal antibodies (mAbs) directed against CLCF1 is an effective method for detecting human CLCF1 protein expression. A validated methodology employs commercial anti-CLCF1 mAbs that can detect both human and mouse CLCF1. This approach involves cell permeabilization followed by intracellular staining with the specific antibody. The specificity of this detection method has been confirmed using conditional knock-out mouse models, making it reliable for human samples as well. This technique differs from previous detection attempts as it can be performed on both mouse and human cytokines and relies on commercially available monoclonal antibodies .
CLCF1 primarily signals through binding to the ciliary neurotrophic factor receptor (CNTFR), which activates downstream signaling cascades. Upon receptor binding, CLCF1 triggers the signal transducer and activator of transcription 3 (STAT3) signaling pathway. In adipocytes, activated STAT3 transcriptionally inhibits both peroxisome proliferator-activated receptor-γ coactivator (PGC) 1α and 1β, which subsequently restrains mitochondrial biogenesis. Unlike other cytokines such as IL-6, CLCF1 shows specific binding patterns; for instance, while both CLCF1 and IL-6 trigger STAT3 phosphorylation in whole bone marrow cells, CLCF1 fails to induce STAT3 activation in sorted LSK (Lineage-Sca-1+c-Kit+) cells, suggesting indirect effects on certain cell populations .
Mutations in the CLCF1 gene can cause Crisponi/cold-induced sweating syndrome (CS/CISS) in humans. This rare genetic disorder is characterized by noninfectious hyperthermia and other symptoms related to thermoregulatory dysfunction. The syndrome demonstrates the critical role of CLCF1 in human temperature regulation and metabolic homeostasis. The involvement of CLCF1 in this disorder provides valuable insights into the physiological significance of CLCF1 signaling in human thermoregulation and suggests potential therapeutic targets for disorders involving temperature dysregulation .
CLCF1 functions as a negative regulator of thermogenesis in brown adipose tissue by activating the CNTFR-STAT3 signaling pathway. Upon activation, STAT3 transcriptionally inhibits both PGC-1α and PGC-1β expression, which restrains mitochondrial biogenesis in brown adipocytes. This mechanism explains how CLCF1 signaling leads to loss of brown fat identity and impairs thermogenic capacity. Studies using adipocyte-specific CLCF1 transgenic (CLCF1-ATG) mice have demonstrated impaired energy expenditure and severe cold intolerance, with elevated CLCF1 triggering whitening of brown adipose tissue. This process is particularly relevant in obesity, where CLCF1 levels are considerably increased, potentially contributing to the limited thermogenesis observed in obese individuals .
CLCF1 exhibits a myeloid-biased hematopoiesis-stimulating effect. In experimental models, CLCF1 increases the frequency and absolute counts of LSK (Lineage-Sca-1+c-Kit+) cells, particularly multipotent progenitor cells. The effect of CLCF1 on hematopoietic stem cells (HSCs) appears to be indirect, as CLCF1 stimulation of whole bone marrow triggers the upregulation of various hematopoiesis-regulating factors. In congenic bone marrow transplantation models, CLCF1 administration leads to accelerated recovery of circulating myeloid cells, with significant differences observable as early as 2 weeks post-transplantation. Notably, CLCF1-treated mice show higher percentages of chimerism and increased absolute counts of donor LSKs, suggesting enhanced engraftment of progenitor cells. The maintenance of high LSK counts after prolonged exposure to CLCF1 suggests it may restrain LSK cell differentiation, possibly through factors like IL-10 that promote HSC self-renewal .
For isolating and purifying CLCF1 for functional studies, researchers should implement a multi-step process. First, recombinant expression systems using mammalian cell lines (such as HEK293T) transfected with CLCF1 cDNA constructs containing affinity tags (His or FLAG) provide an effective approach. After expression, the protein can be purified using affinity chromatography, followed by size exclusion chromatography to ensure high purity. For validation of the purified protein, Western blotting with specific anti-CLCF1 monoclonal antibodies and functional assays measuring STAT3 phosphorylation in responsive cell lines are essential. When working with native CLCF1 from biological samples, immunoprecipitation techniques using the validated monoclonal antibodies that have been shown to detect CLCF1 by flow cytometry can be employed. Importantly, researchers should verify the bioactivity of purified CLCF1 through cell-based assays before proceeding with functional studies .
CLCF1 functions within a complex network of cytokines during inflammatory and immune responses. When whole bone marrow cells are stimulated with CLCF1, they upregulate various factors known to regulate hematopoiesis in a dose-dependent manner. This cytokine cross-talk is crucial for CLCF1's effects on hematopoietic stem and progenitor cells. For example, conditioned media derived from CLCF1-treated whole bone marrow can expand LSK cells to the same extent as direct CLCF1 stimulation, indicating that CLCF1 mediates its effects through the upregulation of pro- and anti-inflammatory cytokines. Among these factors, IL-10 might be particularly important due to its involvement in promoting HSC self-renewal. Additionally, sustained hematopoiesis has been linked to VEGF activity, which is also influenced by CLCF1 signaling. CLCF1 upregulation has also been associated with T helper (Th) 17 polarization, suggesting intricate interactions with the adaptive immune system .
Studying CLCF1-CNTFR interactions presents several methodological challenges. A primary difficulty is distinguishing between CLCF1 and other cytokines that bind to shared receptor components, particularly since CNTFR also binds CNTF (Ciliary Neurotrophic Factor). To overcome this, researchers have developed affinity-matured soluble receptor decoys (eCNTFR-Fc) that selectively bind CLCF1 over CNTF. This engineering approach has increased specificity towards CLCF1 while decreasing binding to CNTF, allowing for more precise investigation of CLCF1-specific signaling.
Another challenge is the species-specific nature of cytokine-receptor interactions. Importantly, mouse CLCF1 can activate CNTFR in human cells, making cross-species studies feasible. For competition binding assays to measure the effect of receptor decoys on CLCF1-receptor complex formation, careful design is necessary to ensure physiological relevance.
When investigating downstream signaling, researchers must consider the indirect effects of CLCF1 on certain cell populations. For example, while both CLCF1 and IL-6 trigger STAT3 phosphorylation in whole bone marrow cells, CLCF1 fails to induce STAT3 activation in sorted LSK cells. This necessitates careful experimental design that accounts for potential mediating factors in CLCF1 signaling pathways .
To differentiate between direct and indirect effects of CLCF1 on target cells, researchers should implement multi-component experimental designs. One effective approach is a conditioned media transfer experiment, where whole bone marrow (WBM) cells are first primed with CLCF1, washed, and then cultured for an additional period. The resulting conditioned media is then transferred to freshly isolated target cells to assess indirect effects. This method revealed that conditioned media derived from CLCF1-treated WBM could expand LSK cells to the same extent as direct CLCF1 stimulation, demonstrating its indirect effect.
Complementary to this, phosphoflow cytometry can be used to examine direct signaling activation. By stimulating either WBM or FACS-sorted target cell populations (such as LSKs) with CLCF1 and measuring STAT3 phosphorylation, researchers can determine if CLCF1 directly activates signaling in specific cell types. For instance, studies have shown that CLCF1 triggers STAT3 phosphorylation in WBM but fails to do so in sorted LSK cells.
Comprehensive analysis of secreted factors using cytokine arrays on supernatants collected from CLCF1-stimulated cells can identify mediating factors. This approach has revealed that CLCF1 triggers the upregulation of various hematopoiesis-regulating factors in a dose-dependent manner, explaining its indirect effects on target cells .
Analysis of CLCF1 expression data in clinical samples requires careful statistical consideration due to its non-normal distribution patterns. Studies have shown that CLCF1 mRNA and protein data often do not follow normal distribution, though CLCF1 protein data can follow normal distribution after log-transformation. Therefore, appropriate statistical methods must be selected based on data distribution.
For normally distributed data (such as log-transformed CLCF1 protein levels), parametric tests like t-tests (for comparing two groups) and one-way ANOVA (for comparing three or more groups) with post-hoc analysis using the least significant difference (LSD) method are appropriate. For non-normally distributed data, non-parametric tests should be employed: Mann-Whitney U test for comparing two groups and Kruskal-Wallis H test followed by LSD post hoc tests for pairwise comparisons among three or more groups.
When analyzing relationships between CLCF1 and clinical parameters such as fractures, hypertension, or coronary heart disease, binary univariate logistic regression (Enter method) is appropriate. Variables with significant differences in univariable analyses should then be incorporated into multiple logistic regression analysis (forward-LR method). Correlation analysis between CLCF1 and continuous clinical variables like bone mineral density can be performed using Pearson (for normally distributed data) or Spearman (for non-normally distributed data) correlation tests .
Optimization of engineered decoy receptors targeting CLCF1 for therapeutic applications involves several critical considerations. First, binding selectivity is essential—engineering decoy receptors like eCNTFR-Fc to preferentially bind CLCF1 over CNTF minimizes potential side effects from inhibiting CNTF signaling, which is important for neuronal cell survival. This selectivity can be achieved through affinity maturation techniques, resulting in increased specificity towards CLCF1 and decreased binding to CNTF.
Cross-species reactivity is another important factor. For preclinical testing, engineered decoy receptors should bind both human and mouse CLCF1 with high affinity. The eCNTFR-Fc has demonstrated this capability, binding mouse CLCF1 with high affinity compared to wild-type CNTFR-Fc.
Functional validation through competition binding assays is crucial to ensure the decoy receptor can effectively sequester CLCF1 and block receptor complex formation. Additionally, stability optimization and pharmacokinetic studies are necessary to evaluate the half-life and tissue distribution of the engineered receptor.
Target population selection is also critical, as evidenced by the observation that CLCF1 blockade appears most effective in tumors driven by oncogenic KRAS, particularly mutations that retain the intrinsic capacity to hydrolyze GTP. This suggests that patient stratification based on molecular markers may be necessary for effective therapeutic application .
Current CLCF1 research faces several limitations that define future research directions. A significant challenge is the complexity of CLCF1 signaling networks across different tissue types. While CLCF1's role in adipose tissue thermogenesis and hematopoiesis has been investigated, its functions in other tissues remain poorly understood. Future research should expand to comprehensive tissue-specific knockout models to elucidate the full spectrum of CLCF1 functions.
The dual autocrine and paracrine signaling mechanisms of CLCF1, particularly in cancer microenvironments, present another challenge. LUAD cell lines and cancer-associated fibroblasts both secrete CLCF1, suggesting complex signaling patterns that require further investigation using co-culture systems and in vivo models.
Mechanistically, the relationship between CLCF1 and KRAS mutations in cancer progression remains incompletely understood. The observation that CLCF1 blockade is most effective in tumors with KRAS mutations that retain GTP hydrolysis capacity suggests a specific interaction that warrants deeper investigation.
Translational research opportunities include exploring CLCF1's potential in ex-vivo hematopoietic stem cell expansion for clinical applications, given its ability to promote LSK proliferation while maintaining functionality. Additionally, the development of more specific inhibitors beyond decoy receptors, such as small molecule antagonists of CLCF1-CNTFR interaction, could provide new therapeutic approaches for conditions ranging from metabolic disorders to cancer .
Parameter | KRAS Mutant | KRAS Wild-type | Statistical Significance |
---|---|---|---|
Cox Hazard Ratio for High CLCF1 Expression | 2.53 (95% CI 1.43–4.48) | 0.86 (95% CI 0.51–1.4) | P=0.001 vs P=0.56 |
CAFs with Elevated CLCF1 vs Matched NLFs | 6 out of 8 patients | N/A | Significant elevation |
Therapeutic Response to eCNTFR-Fc | Effective (especially in G12C) | Less effective | Correlation with GTP hydrolysis capacity |
This data table summarizes the differential impact of CLCF1 expression in KRAS-mutant versus wild-type lung adenocarcinoma, highlighting its potential as a targeted therapeutic approach specifically for KRAS-mutant tumors .
Parameter | Control Condition | CLCF1 Treatment | Effect |
---|---|---|---|
LSK Frequency (24h) | Baseline | Increased | Significant expansion |
LSK Functionality | Normal colony formation | Increased colony formation | Enhanced stem cell activity |
Circulating Myeloid Cell Recovery (post-BMT) | Normal rate (4 weeks) | Accelerated (2 weeks) | Faster reconstitution |
Chimerism Percentage (4 weeks post-BMT) | Baseline | Higher percentage | Enhanced engraftment |
Donor LSK Maintenance | Normal levels | Elevated at weeks 4 and 9 | Sustained hematopoiesis |
This table illustrates the effects of CLCF1 on hematopoietic stem and progenitor cells, demonstrating its potential application in enhancing bone marrow transplantation and hematopoietic recovery .
Method | Application | Advantages | Limitations |
---|---|---|---|
Flow Cytometry with mAb | Detection of cellular CLCF1 | Applicable to both human and mouse CLCF1; Commercial mAb available | Requires cell permeabilization |
Cytokine Arrays | Analysis of CLCF1-induced factors | Broad spectrum analysis; Dose-dependent effects | Semi-quantitative |
Statistical Analysis for Clinical Samples | CLCF1 expression correlation with clinical parameters | Appropriate for non-normal distribution after log-transformation | Requires careful selection of statistical tests |
Engineered Decoy Receptor Binding Assays | Measurement of CLCF1-receptor interactions | Can assess binding specificity and affinity | May not fully reflect in vivo complexity |
This methodological table provides researchers with a comparison of different approaches for detecting and analyzing CLCF1 in experimental and clinical settings .
Parameter | Normal Condition | Obesity | CLCF1 Transgenic Model |
---|---|---|---|
CLCF1 Levels | Decrease during thermogenic stimulation | Considerably increased | Artificially elevated |
Brown Adipose Tissue Identity | Maintained | Compromised | Whitening of brown adipose tissue |
Energy Expenditure | Normal | Reduced | Impaired |
Cold Tolerance | Normal | Reduced | Severe intolerance |
Mitochondrial Biogenesis | Active | Suppressed | Suppressed |
PGC-1α and PGC-1β Expression | Normal | Inhibited | Transcriptionally inhibited |
This table summarizes the role of CLCF1 in regulating brown adipose tissue function and the metabolic consequences of its dysregulation in obesity and transgenic models .
Property | CLCF1 | CNTF | Implications |
---|---|---|---|
Primary Receptor | CNTFR | CNTFR and IL-6R | CNTF has broader receptor binding |
Binding to wtCNTFR-Fc | High affinity | High affinity | Both cytokines recognized by wild-type receptor |
Binding to eCNTFR-Fc | High affinity | No binding | Engineered receptor is CLCF1-specific |
Cross-species Activity | Mouse CLCF1 activates human CNTFR | Species-specific | Facilitates preclinical to clinical translation |
Downstream Signaling | STAT3 activation in certain cell populations | Broader STAT3 activation | Cell-type specific responses |
This comparative table highlights the differences between CLCF1 and CNTF in terms of receptor binding and signaling, which has important implications for developing targeted therapeutic approaches .
CLCF1 forms a heterodimeric complex with Cytokine Receptor-Like Factor 1 (CRLF1) . This complex competes with Ciliary Neurotrophic Factor (CNTF) for binding to the Ciliary Neurotrophic Factor Receptor (CNTFR) complex, thereby activating the Jak-STAT signaling cascade . The CLCF1-CRLF1 complex can be actively secreted from cells by forming a complex with soluble type I CRLF1 or soluble CNTFR .
Mutations in the CLCF1 gene are associated with Crisponi/Cold-Induced Sweating Syndrome 2 (CISS2) . This syndrome is characterized by profuse sweating after exposure to cold and congenital physical abnormalities of the head and spine . The CLCF1-CRLF1 complex has also been implicated in various developmental and pathological conditions, highlighting its importance in both health and disease .